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:

Functional landscapes of POLE and POLD1 mutations in checkpoint blockade-dependent antitumor immunity

Abstract

Defects in pathways governing genomic fidelity have been linked to improved response to immune checkpoint blockade therapy (ICB). Pathogenic POLE/POLD1 mutations can cause hypermutation, yet how diverse mutations in POLE/POLD1 influence antitumor immunity following ICB is unclear. Here, we comprehensively determined the effect of POLE/POLD1 mutations in ICB and elucidated the mechanistic impact of these mutations on tumor immunity. Murine syngeneic tumors harboring Pole/Pold1 functional mutations displayed enhanced antitumor immunity and were sensitive to ICB. Patients with POLE/POLD1 mutated tumors harboring telltale mutational signatures respond better to ICB than patients harboring wild-type or signature-negative tumors. A mutant POLE/D1 function-associated signature-based model outperformed several traditional approaches for identifying POLE/POLD1 mutated patients that benefit from ICB. Strikingly, the spectrum of mutational signatures correlates with the biochemical features of neoantigens. Alterations that cause POLE/POLD1 function-associated signatures generate T cell receptor (TCR)-contact residues with increased hydrophobicity, potentially facilitating T cell recognition. Altogether, the functional landscapes of POLE/POLD1 mutations shape immunotherapy efficacy.

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: Mouse tumors harboring Pole/d1 functional mutations are sensitive to immunotherapy.
Fig. 2: Immune microenvironment of tumors harboring PoleP286R functional mutations.
Fig. 3: Dissecting the de novo mutational signatures in the B16F10 PoleP286R model.
Fig. 4: Statistical models based on mutational signatures can accurately identify tumors harboring POLE/D1 functional mutations from WES data and target panel sequencing data.
Fig. 5: POLE/D1 functional mutation/signature-positive tumors are more immune active and share similar immune features with the baseline mouse PoleP286R tumors.
Fig. 6: Patients with POLE/D1 functional mutations/signatures have better response and survival after anti-PD-1/PD-L1 immunotherapy.
Fig. 7: POLE/D1 function-associated signatures positive status is an independent predictor that can enrich patients who benefit from immunotherapy in the patient population with POLE/D1 mutation.
Fig. 8: Trinucleotide context spectrum of SBS mutational signatures and immunogenicity of neoantigens.

Similar content being viewed by others

Data availability

Mutation data from the TCGA pancancer study (mc3.v0.2.8.PUBLIC.maf.gz) were downloaded from NCI genomic data commons (https://api.gdc.cancer.gov/data/1c8cfe5f-e52d-41ba-94da-f15ea1337efc). Mutation data of ICGC and CCLE were downloaded from the ICGC data portal (https://dcc.icgc.org/api/v1/download?fn=/current/Summary/simple_somatic_mutation.aggregated.vcf.gz) and the Broad Institute website (https://ndownloader.figshare.com/files/34008434), respectively. To generate the ICGC/CCLE test set, TCGA samples in the ICGC cohort were removed manually. BED files of MSK-IMPACT sequencing regions are acquired from AACR-Genie project database (https://www.synapse.org/#!Synapse:syn7222066/wiki/405659). COSMIC SBS exome signature v.3 and COSMIC SBS exome TSB signature v.3 were downloaded from the Synapse database (https://www.synapse.org/#!Synapse:syn11967914). The mouse GRCm38 genome was download from UCSC genome browser (https://hgdownload.soe.ucsc.edu/goldenPath/mm10/bigZips/mm10.fa.gz). A baseline list of known POLE/D1 functional mutations was generated from previous publications and the OncoKB database (https://www.oncokb.org/)12,35. Additional lists of clinical associated POLE/D1 mutations and POLE/D1 mutator alleles in other species were curated manually from the literature and are available as Supplementary Tables 2 and 3. Gene expression profiles and xCell-based cell type deconvolutions of TCGA-endometrial cohort were downloaded from the TIMER database (http://timer.cistrome.org/infiltration_estimation_for_tcga.csv.gz)85,86. TCRb repertoire data were downloaded from NCI genomic data commons (TCGA_mitcr_cdr3_result_161008.tsv, from https://gdc.cancer.gov/about-data/publications/panimmune)87.TCGA predicted neoantigen information was acquired from TSNAdb (http://biopharm.zju.edu.cn/tsnadb/download/)88. Mouse WES and RNA-seq data were deposited to SRA (PRJNA701709). All the other data were deposited with Synapse (as Synapse project syn29404148, https://www.synapse.org/#!Synapse:syn29404148/wiki/617361) with public access. All samples underwent evaluation for model building (Synapse ID: syn29477489.1), the mutational signature matrix for training and test the WES (Synapse ID: syn29478035, Synapse ID: syn29478033) and MSK-IMPACT models (Synapse ID: syn29478110, Synapse ID: syn29478151), the genomic, response and survival data of the MSK-IMPACT immunotherapy cohorts (Synapse ID: syn29478036).

Code availability

All customized code including code for generating the models (Synapse ID: syn29479495)89, analysis of the MSK-IMPACT cohort (Synapse ID: syn29479497)90 and other associated code (Synapse ID: syn30137113)91 have been deposited with Synapse (Synapse project ID: syn29404148, https://www.synapse.org/#!Synapse:syn29404148/wiki/617361) with public access. Code for processing WES and RNA-seq was from published tools and is available from the authors of the tools as described in Methods.

References

  1. Borcoman, E. et al. Novel patterns of response under immunotherapy. Ann. Oncol. 30, 385–396 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Hegde, P. S. & Chen, D. S. Top 10 challenges in cancer immunotherapy. Immunity 52, 17–35 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Mouw, K. W., Goldberg, M. S., Konstantinopoulos, P. A. & D’Andrea, A. D. DNA damage and repair biomarkers of immunotherapy response. Cancer Discov. 7, 675–693 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Bever, K. M. & Le, D. T. DNA repair defects and implications for immunotherapy. J. Clin. Invest. 128, 4236–4242 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  5. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  6. 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 

  7. 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 (2021).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. 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 

  10. Ying, J. et al. Additive effects of variants of unknown significance in replication repair-associated DNA polymerase genes on mutational burden and prognosis across diverse cancers. J. Immunother. Cancer 9, e002336 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Lujan, S. A., Williams, J. S. & Kunkel, T. A. DNA polymerases divide the labor of genome replication. Trends Cell Biol. 26, 640–654 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Campbell, B. B. et al. Comprehensive analysis of hypermutation in human cancer. Cell 171, 1042–1056 e10 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kunkel, T. A. Evolving views of DNA replication (in)fidelity. Cold Spring Harb. Symp. Quant. Biol. 74, 91–101 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Esteban-Jurado, C. et al. POLE and POLD1 screening in 155 patients with multiple polyps and early-onset colorectal cancer. Oncotarget 8, 26732–26743 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Mehnert, J. M. et al. Immune activation and response to pembrolizumab in POLE-mutant endometrial cancer. J. Clin. Invest. 126, 2334–2340 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  16. van Gool, I. C. et al. POLE proofreading mutations elicit an antitumor immune response in endometrial cancer. Clin. Cancer Res. 21, 3347–3355 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Domingo, E. et al. Somatic POLE proofreading domain mutation, immune response, and prognosis in colorectal cancer: a retrospective, pooled biomarker study. Lancet Gastroenterol. Hepatol. 1, 207–216 (2016).

    Article  PubMed  Google Scholar 

  18. 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 

  19. He, J. et al. Distinctive genomic characteristics in POLE/POLD1-mutant cancers can potentially predict beneficial clinical outcomes in patients who receive immune checkpoint inhibitor. Ann. Transl. Med. 9, 129 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Barbari, S. R. & Shcherbakova, P. V. Replicative DNA polymerase defects in human cancers: consequences, mechanisms, and implications for therapy. DNA Repair (Amst.) 56, 16–25 (2017).

    Article  CAS  PubMed Central  Google Scholar 

  21. Silberman, R. et al. Complete and prolonged response to immune checkpoint blockade in POLE-Mutated colorectal cancer. JCO Precis. Oncol. 3, 1–5 (2019).

    PubMed  Google Scholar 

  22. Rayner, E. et al. A panoply of errors: polymerase proofreading domain mutations in cancer. Nat. Rev. Cancer 16, 71–81 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature 578, 94–101 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Petljak, M. et al. Characterizing mutational signatures in human cancer cell lines reveals episodic APOBEC mutagenesis. Cell 176, 1282–1294 e20 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Koh, G., Degasperi, A., Zou, X., Momen, S. & Nik-Zainal, S. Mutational signatures: emerging concepts, caveats and clinical applications. Nat. Rev. Cancer 21, 619–637 (2021).

    Article  CAS  PubMed  Google Scholar 

  26. Albertson, T. M. et al. DNA polymerase epsilon and delta proofreading suppress discrete mutator and cancer phenotypes in mice. Proc. Natl Acad. Sci. USA 106, 17101–17104 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Li, H. D. et al. Polymerase-mediated ultramutagenesis in mice produces diverse cancers with high mutational load. J. Clin. Invest. 128, 4179–4191 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  30. Knijnenburg, T. A. et al. Genomic and molecular landscape of DNA damage repair deficiency across the Cancer Genome Atlas. Cell Rep. 23, 239–254 e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Leon-Castillo, A. et al. Interpretation of somatic POLE mutations in endometrial carcinoma. J. Pathol. 250, 323–335 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bellido, F. et al. POLE and POLD1 mutations in 529 kindred with familial colorectal cancer and/or polyposis: review of reported cases and recommendations for genetic testing and surveillance. Genet. Med. 18, 325–332 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Aran, D., Hu, Z. & Butte, A. J. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 18, 220 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Chakravarty, D. et al. OncoKB: a precision oncology knowledge base. JCO Precis. Oncol., doi:DOI: 10.1200/PO.17.00011 (2017)..

  36. Hoadley, K. A. et al. Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell 173, 291–304 e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Youden, W. J. Index for rating diagnostic tests. Cancer 3, 32–35 (1950).

    Article  CAS  PubMed  Google Scholar 

  38. International Cancer Genome, C. et al. International network of cancer genome projects. Nature 464, 993–998 (2010).

    Article  Google Scholar 

  39. Ghandi, M. et al. Next-generation characterization of the cancer cell line encyclopedia. Nature 569, 503–508 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. 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 

  41. Chowell, D. et al. Improved prediction of immune checkpoint blockade efficacy across multiple cancer types. Nat. Biotechnol. 40, 499–506 (2022).

    Article  CAS  PubMed  Google Scholar 

  42. Chowell, D. et al. TCR contact residue hydrophobicity is a hallmark of immunogenic CD8+ T cell epitopes. Proc. Natl Acad. Sci. USA 112, E1754–E1762 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Riley, T. P. et al. Structure based prediction of neoantigen immunogenicity. Front Immunol. 10, 2047 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Calis, J. J. et al. Properties of MHC class I presented peptides that enhance immunogenicity. PLoS Comput. Biol. 9, e1003266 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Li, H. D. et al. A PoleP286R mouse model of endometrial cancer recapitulates high mutational burden and immunotherapy response. JCI Insight 5, e138829 (2020).

    Article  PubMed Central  Google Scholar 

  47. Davies, H. et al. HRDetect is a predictor of BRCA1 and BRCA2 deficiency based on mutational signatures. Nat. Med. 23, 517–525 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lee, J. S. & Ruppin, E. Multiomics prediction of response rates to therapies to inhibit programmed cell death 1 and programmed cell death 1 ligand 1.JAMA Oncol. 5, 1614–1618 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. 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).

    Article  CAS  PubMed  Google Scholar 

  50. Litchfield, K. et al. Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell 184, 596–614 e14 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. van Elsas, A., Hurwitz, A. A. & Allison, J. P. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J. Exp. Med. 190, 355–366 (1999).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Zitvogel, L., Pitt, J. M., Daillere, R., Smyth, M. J. & Kroemer, G. Mouse models in oncoimmunology. Nat. Rev. Cancer 16, 759–773 (2016).

    Article  CAS  PubMed  Google Scholar 

  53. Zeng, Z. et al. TISMO: syngeneic mouse tumor database to model tumor immunity and immunotherapy response. Nucleic Acids Res. 50, D1391–D1397 (2022).

    Article  CAS  PubMed  Google Scholar 

  54. Griswold, D. P. & Corbett, T. H. A colon tumor model for anticancer agent evaluation. Cancer 36, 2441–2444 (1975).

    Article  CAS  PubMed  Google Scholar 

  55. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Clement, K. et al. CRISPResso2 provides accurate and rapid genome editing sequence analysis. Nat. Biotechnol. 37, 224–226 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 20, 1297–1303 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. 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 

  60. Koboldt, D. C. et al. VarScan: variant detection in massively parallel sequencing of individual and pooled samples. Bioinformatics 25, 2283–2285 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Kim, S. et al. Strelka2: fast and accurate calling of germline and somatic variants. Nat. Methods 15, 591–594 (2018).

    Article  CAS  PubMed  Google Scholar 

  62. Larson, D. E. et al. SomaticSniper: identification of somatic point mutations in whole genome sequencing data. Bioinformatics 28, 311–317 (2012).

    Article  CAS  PubMed  Google Scholar 

  63. McLaren, W. et al. The Ensembl variant effect predictor. Genome Biol. 17, 122 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  65. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Rosenbloom, K. R. et al. The UCSC Genome Browser database: 2015 update. Nucleic Acids Res. 43, D670–D681 (2015).

    Article  CAS  PubMed  Google Scholar 

  67. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Reich, M. et al. GenePattern 2.0. Nat. Genet. 38, 500–501 (2006).

    Article  CAS  PubMed  Google Scholar 

  69. Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  71. 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 

  72. Bergstrom, E. N. et al. SigProfilerMatrixGenerator: a tool for visualizing and exploring patterns of small mutational events. BMC Genomics 20, 685 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Blokzijl, F., Janssen, R., van Boxtel, R. & Cuppen, E. MutationalPatterns: comprehensive genome-wide analysis of mutational processes. Genome Med 10, 33 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Rosenthal, R., McGranahan, N., Herrero, J., Taylor, B. S. & Swanton, C. DeconstructSigs: delineating mutational processes in single tumors distinguishes DNA repair deficiencies and patterns of carcinoma evolution. Genome Biol. 17, 31 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  75. 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 

  76. 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 

  77. 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 

  78. 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 

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

    Article  PubMed  PubMed Central  Google Scholar 

  80. Schwartz, L. H. et al. RECIST 1.1-Update and clarification: from the RECIST committee. Eur. J. Cancer 62, 132–137 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Liu, X., Li, C., Mou, C., Dong, Y. & Tu, Y. dbNSFP v4: a comprehensive database of transcript-specific functional predictions and annotations for human nonsynonymous and splice-site SNVs. Genome Med 12, 103 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    Article  CAS  PubMed  Google Scholar 

  83. Kyte, J. & Doolittle, R. F. A simple method for displaying the hydropathic character of a protein. J. Mol. Biol. 157, 105–132 (1982).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  85. Li, B. et al. Comprehensive analyses of tumor immunity: implications for cancer immunotherapy. Genome Biol. 17, 174 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  86. Sturm, G. et al. Comprehensive evaluation of transcriptome-based cell-type quantification methods for immuno-oncology. Bioinformatics 35, i436–i445 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Wu, J. et al. TSNAdb: a database for tumor-specific neoantigens from immunogenomics data analysis. Genomics Proteom. Bioinforma. 16, 276–282 (2018).

    Article  Google Scholar 

  89. Ma, X. et al. NG_WES_IMPACT_Model_training_testing_script.R [R code]. Synapse https://doi.org/10.7303/syn29479497.1 (2022).

  90. Ma, X. et al. NG_ICB_Cohort_script.R [R code]. Synapse https://doi.org/10.7303/syn29479495 (2022).

  91. Ma, X. et al. NG-A57433 other associated codes.R [R code]. Synapse https://doi.org/10.7303/syn30137113 (2022).

Download references

Acknowledgements

We thank colleagues at the Cleveland Clinic flow core, Cleveland Clinic genomics core, Center for Immunotherapy and Precision-Immuno-Oncology Shared Platforms, MSK core facilities (including Integrated Genomics Operation (IGO), Flow Cytometry Core Facility (FCCF) and Molecular Cytology Core Facility (MCCF)) for processing our samples and providing important suggestions. We thank colleagues at the Molecular Diagnostics Service in the Department of Pathology, and the Marie-Josee and Henry R. Kravis Center for Molecular Oncology Marie-Josée of MSK for establishing and generating the MSK-IMPACT data. We thank all the members and alumni of the Chan laboratory at MSK and CCF, as well as the LRI of CCF and HOPP of MSK for their generous help and support of this study. The results presented here are based in part on data generated or collected by the TCGA Research Network, Broad CCLE, ICGC. We acknowledge funding sources including National Institutes of Health (NIH) R01 CA205426 (T.A.C.), NIH R35 CA232097 (T.A.C.), the STARR Cancer Consortium (T.A.C.), NIH DP5 OD028171 (R.M.S), the Burroughs Wellcome Fund (R.M.S.) and the NIH/National Cancer Institute Cancer Center Support Grant P30 CA008748 (MSKCC).

Author information

Authors and Affiliations

Authors

Contributions

X.M. and T.A.C. conceptualized the study. X.M., V.M., F.K., D.H., N.R. and T.A.C. developed the methodology. X.M., V.M, F.K., R.M.S., D.H., F.K., H.J., C.W.R.F., J.A., P.B.P., Y.Z., L.K.C., S.L.W., D.C., E.Y.S., M.L., J.W., R.V., L.V., W.Y., X.Z., C.V., L.G.T.M., N.R. and T.A.C. performed the investigaion. X.M., V.M., F.K., D.H., N.R. and T.A.C. wrote the original draft of the manuscript. X.M., R.M.S., D.C., T.J.A., I.J., L.G.T.M., N.R. and T.A.C. reviewed and edited the manuscript. M.G. carried out the biostatistical review. T.A.C. acquired funding. T.A.C. supervised the project. All authors critically discussed and revised the manuscript for important intellectual content.

Corresponding author

Correspondence to Timothy A. Chan.

Ethics declarations

Competing interests

T.A.C. is a cofounder 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., R.M.S. and D.C. are inventors on intellectual property held by MSKCC on using tumor mutation burden to predict immunotherapy response, with pending patent, which has been licensed to PGDx. C.V. acknowledges research grant funding from Fundación Alfonso Martín Escudero. D.Z. received consulting fees from Agenus, Hookipa Biotech, Targovax, AstraZeneca, Synthekine, Mana Therapeutics, Xencor, Crown Biosciences and Memgen. D.Z. receives grant/research support from AstraZeneca, Roche and Plexxikon. D.Z. holds stock options for Immunos Therapeutics, Calidi Biotherapeutics, Mana Therapeutics and Accurius. D.Z. has a patent related to use of Newcastle Disease Virus for cancer therapy with royalties paid from Merck. R.Y. has served as an advisor for Natera, Array BioPharma/Pfizer and Mirati Therapeutics and has received research support to her institution from Array BioPharma/Pfizer, Boehringer Ingelheim and Mirati Therapeutics. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Shiaw-Yih Lin 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 Mouse tumors harboring Pole/d1 functional mutations are sensitive to immunotherapy.

a, Strategy to introduce PoleP286R into murine cell lines. b, Scheme of in vitro culture and WES (whole exome sequencing) of parental and PoleP286R mutant cell lines. c, Total SNV (single nucleotide variant) counts of the PoleP286R mutant and parental cell lines. P values by two-sided Student’s t-tests. d, B16F10 PoleP286R clone2 cell line with ICB therapies (N = 15 mice per group). P values (anti-CTLA4, P = 0.0003; anti-PD1, P = 0.0002; Combo, P = 0.0001). e, Survival analysis of mice bearing the B16F10 parental (anti-CTLA4, P = 0.14; anti-PD1, P = 0.13; Combo, P = 0.0003), the B16F10 PoleP286R (anti-CTLA4, P < 0.0001; anti-PD1, P < 0.0001; Combo, P < 0.0001) or the B16F10 PoleP286R clone2 tumors (anti-CTLA4, P < 0.0001; anti-PD1, P < 0.0001; Combo, P < 0.0001) after ICB (N = 15 mice per group). P values indicate log-rank test significance. f, CT-26- PoleP286R clone2 cell line with ICB therapies (N = 15 mice per group). P values (CTLA4 P = 1.1e-8, PD-1 P = 0.0015, Combo P = 3.2e-9). g, CT-26 PoleWT single cell clone1&2 tumors with ICB therapy (N = 15 mice per group). P values (Clone1 P = 0.023, Clone2 P = 0.053). h, Tumor inhibition rates of the CT-26 parental, PoleWT single clones and PoleP286R clone1 and clone2 tumors with anti-PD1 therapy at the last time point. (N = 15). Dots represent individual biological replicates. P values by two-sided Student’s t-tests. i, Growth curves of the B16F10 PoleV411L clone2 with anti-PD1 therapy (N = 15 mice per group, P = 0.0001). For all growth curves related panels (d,f,g,i), P values by two-sided Student’s t-tests at the end time points. For all panels, data are presented as mean values ± s.e.m. (n.s., no statistical significance, * P < 0.05, ** P < 0.01, *** P < 0.005, **** P < 0.0001). No multiple comparisons adjustment was performed.

Extended Data Fig. 2 The baseline immune microenvironment of the Pole mutant tumors.

a, Heatmap of 1298 DEGs (differentially regulated genes) from RNA-seq analysis of the B16F10 parental and PoleP286R mutant tumors 14 days post implantation. Color sale indicates normalized z-score. b, GSEA (gene set enrichment assay) indicating enrichment of gene sets related to interferon gamma response, T cell and NK cell activation, inflammation, antigen presenting pathway and PD1 signaling in the mutant tumors versus parental tumors. c, Heatmap showing DEGs (FDR P < = 0.05) between parental and mutant tumors from the Hallmark interferon gamma response pathway, PID CD8 TCR pathway and KEGG natural killer cell mediated cytotoxicity pathway. d, Ptprc TPM of parental and mutant tumors from the RNA-seq data showed in Fig. 2c (N = 3 biological replicates). P = 0.0008, two-sided Student’s t-tests (n.s., no statistical significance, * P < 0.05, ** P < 0.01, *** P < 0.005). Data are presented as mean values + /- SEM. e, Flow cytometry analysis of the percentage of Cd8 T cells expressing Pd-1 (P = 0.026), Tigit (P = 0.48) and Lag3 (P = 0.78) in the parental and mutant tumors 14 days post implantation (N = 6 biological replicates). P values by two-sided Student’s t-tests (n.s., no statistical significance, * P < 0.05, ** P < 0.01, *** P < 0.005). Data are presented as mean values + /- SEM. f, Flow cytometry analysis of expression intensity of Pd-l1 (P = 0.045), Cd204 (P = 0.043) and Cd206 (P = 0.0087) on tumor associated macrophages in the parental and mutant tumors 14 days post implantation (N = 6 biological replicates). P values by two-sided Student’s t-tests at the end time points (n.s., no statistical significance, * P < 0.05, ** P < 0.01, *** P < 0.005). Data are presented as mean values + /- s.e.m.

Extended Data Fig. 3 Immune microenvironment of the post-treatment Pole mutant tumors.

a, Differentially expressed genes up-regulated or down-regulated in the post-ICB PoleP286R tumors versus the parental tumors. There are 82 DEGs that are consistently upregulated in all ICB treatment arms, while there are 59 genes are consistently downregulated in mutant tumors of all immune checkpoint blockade (ICB) arms. b, Top 10 KEGG pathways that were significantly enriched in the consistently up-regulated DEGs. c, Top 10 KEGG pathways that were enriched in the consistently down-regulated DEGs across all ICB arms. No pathway is statistical significantly enriched, as determined by q value <0.05. d, GSEA analysis of PoleP286R tumor in the combo arm versus the IgG arm. Only pathways with nominal p value <0.05 were shown. e, GSEA plot of enriched gene sets related with inflammatory response in combo ICB arm versus IgG arm of the PoleP286R tumors.

Extended Data Fig. 4 Both adaptive and innate immune cell types contribute to the distinct immune profiles of the post-treatment Pole mutant tumors, compared to that of the parental tumors.

a, Heatmap of immune-cell-type-signatures enrichment in the post-ICB samples. Color scale indicates normalized enrichment scores. b, Screen plot of the principal component analysis. Bars indicate the explained variations for each PC. The red line and dots indicate the accumulatively explained variations from PC1 to each other PC. c, Contribution of each immune-cell-type-signature to PC1. Red indicates the enrichment score of the immune-cell-type-signatures aligned to the same direction with the PC1 axle, blue indicates the enrichment score of the immune-cell-type-signatures aligned to the opposite direction with the PC1 axle. d, Normalized enrichment score of monocytes and macrophages in post-ICB tumors (N = 3 biological replicates). P values (monocytes P = 3.98e-5; macrophage P = 4.1e-6) were derived from two-way ANOVA test. e, Normalized enrichment score of Treg (regulatory T cell) and NKT cells in post-immunotherapy tumors (N = 3 biological replicates). P values (Treg P = 0.030; NKT P = 0.022) were derived from two-way ANOVA test. i, z-score transformed fraction of the TCR-beta CDR3 clone types in the post-treated parental and PoleP286R tumors. Note that no CDR3 clone type is successfully extracted from two of the parental tumors treated with IgG. f, Chao1 index and richness score of the TCR-beta CDR3 sequence of post-treated tumors (N = 3 biological replicates). g-h, Chao1, richness, evenness and clonality scores of the TCR-beta CDR3 sequences of post-treated tumors (N = 3 biological replicates). P values (Chao1 P = 0.012; Richness P = 0.051; Evenness P = 0.26; Clonality P = 0.26) were derived from two-way ANOVA test. For all boxplots (d-e, g-h), the minima (0% percentile), maxima (100% percentile) were plotted as the whiskers, 25% percentile and 75% percentile were plotted as the bounds of the boxes, medians were plotted as the center bar.

Extended Data Fig. 5 Mutation signatures of the Pole mutant cell lines.

a, Schematic explanation of the baseline and de novo SNVs in parental and mutant cell lines. b, Transcriptional strand bias (TSB) of the six base substitution categories of the baseline and de novo mutations from the B16F10 parental and PoleP286R mutant cell lines. De novo mutations in the parental and the PoleP286R mutation cell lines showed distinct transcriptional strand bias, indicating that these mutations are generated by different biological processes. c, The 192 TSB base substitutions in tri-nucleotide sequence contexts of the three NMF extracted de novo TSB mutational signatures from the baseline and de novo SNVs of the B16F10 parental and PoleP286R mutant cell lines. d, Contribution of the three NMF extracted de novo TSB mutation signatures to the baseline and de novo mutations in the B16F10 parental and PoleP286R mutant cell lines showed that the TSB-Sig.B is exclusively contributed to the de novo mutations discovered in the B16F10 PoleP286R mutant cell lines. e, Cosine similarity of the de novo mutational TSB signatures with COSMIC TSB-SBS signatures v3. The TSB-Sig.B is highly similar to the POLE/D1 function-assocaited signature TSB-SBS10b (cosine similarity of 0.84). f, COSMIC SBS signatures extracted from B16F10 parental and PoleP286R mutant cell lines by NNLS method. The POLE/D1 function-associated signature SBS10b can only be extracted from the de novo SNVs of the PoleP286R mutant samples.

Extended Data Fig. 6 Statistical models based on function-associated signatures can be used to identify tumors with POLE/D1 functional mutations.

a, TCGA data set summary. Wild-type, tumors are wild-type for POLE/D1; Functional, tumors harboring known POLE/D1 functional mutations; Mutated, tumors with only POLE/D1 VUS; SNV, SNV count by WES sequencing. b, the optimal Youden Index point and corresponding probability cutoff value. c, ICGC/CCLE test set summary. d, Heatmap of the Non-negative least squares (NNLS) extracted COSMIC SBS signatures of false negative predictions. e, Reconstitution accuracy of the non-negative matrix factorization (NMF) extracted signatures on TCGA samples with POLE/D1 functional mutations. f, Cosine similarity of the three NMF extracted mutational signatures to the COSMIC SBS signatures. g-h, Contribution of the three NMF de novo mutational signatures to the samples with known POLE/D1 functional mutations in the training set (g) and test set (h). TMB, SNV count in the exome region by WES sequencing. Functional mutation, whether the sample contain POLE or POLD1 functional mutations. i, Fisher exact test on the MSI status of the true-positive and false negative samples from the training and test sets when MSI status is available. j. Tumor allele frequencies of the POLE/D1 functional mutations from the false negative predictions (FN, N = 5), True-positive predictions (TP, N = 77) with available allele frequencies. P value was calculated with two-sided Wilcoxon Rank Sum Test. k, SNV count distribution of the false positive samples from the WES training set. l, Distribution of the true-positive samples (top) or VUS samples (bottom) that were predicted as samples with functional mutations. The green dash lines indicate cutoff for SNVlow (3.6 SNV/Mb exome), the blue dash lines indicate cutoff for SNVint/hi (10 SNV/Mb exome) and the red dash lines indicate cutoff for SNVhyper (50SNV/Mb exome). m. Unsupervised clustering of the SNVint/hi FP samples from the TCGA training set based on the extracted COSMIC SBS signatures.

Extended Data Fig. 7 Statistical models based on function-associated signatures predict functional mutations and associated immune features.

a, Correlation of the proportion of POLE/D1 function-associated signatures extracted from TCGA WES or IMPACT simulation data. Pearson correlation coefficients were shown. b. Confusion table of the WES model on TCGA-impact data. Pred. wild-type, samples that were predicted as wild-type for POLE/D1 functional mutations; Pred. functional, samples that were predicted harboring POLE/D1 functional mutations. c, Scheme of training a model to identify tumors harboring POLE/D1 functional mutations. d, MSK-IMPACT training set summary. e, Optimal Youden Index point and corresponding probability cutoff. f, Heatmap of the NNLS extracted COSMIC SBS mutational signatures of the false negative predictions from the MSK-IMPACT training set. g, Co-efficiencies of the POLE/D1 functional-associated mutational signatures in the WES and IMPACT models. h. Immune infiltration scores and CYT scores of the samples with known POLE/D1 functional mutations (N = 53) and POLE/D1 variant of unknown significance (VUS) samples with function-associated signatures (N = 7) compared to the POLE/D1 functional mutation/signature-negative tumors (N = 520) from the TCGA-endometrial cohort. VUS samples with function-associated signatures, samples harbor POLE/D1 VUSes and were positive for POLE/D1 function-associated signatures predicted by the function-associated signature-based model, POLE/D1 functional mutation/signature-negative tumors, wild type samples or samples harbor POLE/D1 VUSes and didn’t show POLE/D1 function-associated signature. P values represent two-sided Wilcoxon Rank Sum Test. The minima, maxima were plotted as the whiskers, 25% and 75% percentile were plotted as the bounds of the boxes, medians and mean were plotted as the center bar and black dot. i. Screen plot of the principal component analysis. Bars indicate the explained variations for each PC. The red line and dots indicate the accumulatively explained variations. j, Sample separation plot of the three groups of samples in h based on the first two PCs, P values represent permutational multivariate analysis of variance test. (n.s., no statistical significance, * P < 0.05, ** P < 0.01, *** P < 0.005).

Extended Data Fig. 8 Immune features and the outcome of patients with POLE/D1 functional mutations/signatures.

a, Enrichment scores of the immune cell types of sample populations in Fig. 5d (continued). CD4 Tem P = 0.0042; Th1 P = 6.2e-5; Th2 P = 1.6e-9; Eosinophils P = 0.027; Macrophages P = 4.8e-5; Memory B-cell P = 0.036. b, Log-fold changes of the immune cell type enrichment scores from the human tumors and mouse tumors. Red color indicates cell types that are consistently upregulated or downregulated (P < 0.05) for both human and mouse tumor comparisons. c, Chao1 and clonality of the TCR-beta CDR3 repertoires from the sample population in Fig. 5f. Chao1 P = 8.5e-5, clonality P = 0.045. d, SBS signature profiles of the 24 POLE/D1 functional mutation/signature-positive patients in the ICB cohort. e-f. Comparison of the TMB (e) and copy number alterations (f) between the POLE/D1 functional mutation/signature-positive patients, other POLE/D1 mutated patients, and wild-type patients (N = 24, 148 and 2528 patients). TMB, P = 2.2e-5; P = 0.0021; FGA, P < 2.2e-16; P = 0.0095. g, Kaplan-Meier overall survival plot of the POLE/D1 functional mutation/signature-positive patients versus the histology-matched wild-type patients. Log-Rank P value and hazard ratio shown represents coxph model with cancer-type correction. h, Forest plot of the POLE/D1 functional mutations/signatures in coxph models of overall survival after immunotherapy with cancer type correction for pan-cancer or single cancer type categories that have at least three POLE/D1 functional mutation/signature-positive patients. Horizontal bars represent the 95% confidence interval for the hazard ratios. Each line indicates an individual coxph model. Error bar centers indicate Hazard ratios. i, Estimated tumor size change and mutational signatures composition for the complete responder harbored POLEP286R mutation. For all boxplots (a, c, e), P values by Wilcoxon Rank Sum Test. The minima and maxima were plotted as the whiskers, 25% and 75% were plotted as box bounds, medians were plotted as the center bar. For all tests (n.s., no statistical significance, * P < 0.05, ** P < 0.01, *** P < 0.005).

Extended Data Fig. 9 POLE/D1 function-associated signatures predicts immunotherapy response.

a. Proportion of clinical beneficial cases between the POLE/D1 (POLE or POLD1) functional mutation/signature-positive patients and the histology-matched wild-type patients. Functional mutations/signatures, patients either harbored known POLE/D1 functional mutations, or only harbored POLE/D1 VUSes but were predicted as function-associated signature-positive. b, Kaplan-Meier progression free survival plot of the patients harbored any types of POLE/D1 mutations versus POLE/D1 wild-type patients. c, Kaplan-Meier progression free survival plot of the patient populations in a. d-e, Kaplan-Meier overall survival plot (d) and proportion of clinical beneficial cases (e) of the POLE/D1 functional mutation/signature-positive patients versus the POLE/D1 function-associated signature-negative VUS patients after immunotherapy. f. Proportion of clinical beneficial cases of the FP (false positive) prediction wild-type patients versus the TN (true negative) prediction wild-type patients upon ICB. FP prediction, wild-type patients that were predicted as POLE/D1 functional mutation-positive. TN prediction, wild-type patients that were predicted as wild-type samples. g-h, Kaplan-Meier overall survival (g) and progression free survival plot (h) of FP prediction wild-type patients versus TN prediction wild-type patients. I, A multivariable coxph model comparing the predictive capability of different patient selection strategies on the progression free survival on patients after ICB (N = 1130). Horizontal bars represent the 95% confidence interval of the hazard ratio. Error bar centers indicate hazard ratios. j-k, Kaplan-Meier overall survival plot (j) and progression free survival plot (k) of the POLE/D1 exonuclease domain mutation-positive patients versus the POLE/D1 wild-type patients. l-m, Kaplan-Meier overall survival plot (l) and progression free survival plot (m) of the POLE/D1 functional mutation/signature-positive patients that were not hypermutated, versus the POLE/D1 wild-type patients. For a, e-f, P values, Fisher’s exact t-test. For all Kaplan-Meier plots (b-d, g-h, j-m), Log-Rank P value and hazard ratio shown were calculated from coxph model with cancer type correction.

Extended Data Fig. 10 POLE/D1 function-associated signature-based model predicts ICB outcome and outperforms traditional approaches.

a-b. Kaplan-Meier overall survival (a) and progression free survival plot (b) of the patients with at least one POLE/D1 (POLE or POLD1) mutation classified as damaging mutation by all five in silico algorithms versus all the rest POLE/D1 mutated patients. c, C-index of the progression free survival coxph models generated based on different patient selection strategies and patient population in Fig. 7g. Two-sided P values represent paired student t-tests of coxph model based on POLE/D1 functional mutation/signature-positive against other models without multiple comparison adjustment. d, Multi-variable coxph model of ICB progression free survival for ‘POLE/D1 functional mutation/signature-positive’ and TMB with cancer type correction (N = 1130). Only POLE/D1 functional mutation/signature-positive and TMB are shown in the forest plot, * log-rank P < 0.05. *** log-rank P < 0.005. Error bar indicating 95% CI of the Hazard ratio. e, Kaplan-Meier progression free survival plot of the POLE/D1 functional mutation/signature-positive patients versus the POLE/D1 wild-type patients in the ICB-treated patient cohort with high TMB (TMB > = 10). f, Kaplan-Meier overall survival plot of a random sub-cohort of the POLE/D1 functional mutation/signature-positive patients versus the POLE/D1 wild-type patients with matched median and minimum TMB. g, Kaplan-Meier progression free survival plot of a random sub-cohort of the POLE/D1 functional mutation/signature-positive patients versus the POLE/D1 wild-type patients with matched median and minimum TMB. h-i, Proportion heatmap of the observed association between mutational signatures with each SNV class (h) or each amino acid alteration category (i) in the TCGA pan-cancer cohort. In i, amino acids on the top row are the resulting new allele from the mutation (Post); amino acids on the bottom row are the wild-type allele (Pre). For all Kaplan-Meier plots (a-b, e-g), Log-Rank P value and hazard ratio shown were calculated from coxph model with cancer type correction.

Supplementary information

Supplementary Information

Supplementary Notes 1–16 and Figs. 1–6.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–11.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, X., Riaz, N., Samstein, R.M. et al. Functional landscapes of POLE and POLD1 mutations in checkpoint blockade-dependent antitumor immunity. Nat Genet 54, 996–1012 (2022). https://doi.org/10.1038/s41588-022-01108-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-022-01108-w

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