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:

Cancer cells resistant to immune checkpoint blockade acquire interferon-associated epigenetic memory to sustain T cell dysfunction

Abstract

Prolonged interferon (IFN) signaling in cancer cells can promote resistance to immune checkpoint blockade (ICB). How cancer cells retain effects of prolonged IFN stimulation to coordinate resistance is unclear. We show that, across human and/or mouse tumors, immune dysfunction is associated with cancer cells acquiring epigenetic features of inflammatory memory. Here, inflammatory memory domains, many of which are initiated by chronic IFN-γ, are maintained by signal transducer and activator of transcription (STAT)1 and IFN regulatory factor (IRF)3 and link histone 3 lysine 4 monomethylation (H3K4me1)-marked chromatin accessibility to increased expression of a subset of IFN-stimulated genes (ISGs). These ISGs include the RNA sensor OAS1 that amplifies type I IFN (IFN-I) and immune inhibitory genes. Abrogating cancer cell IFN-I signaling restores anti-programmed cell death protein 1 (PD1) response by increasing IFN-γ in immune cells, promoting dendritic cell and CD8+ T cell interactions, and expanding T cells toward effector-like states rather than exhausted states. Thus, cancer cells acquire inflammatory memory to augment a subset of ISGs that promote and predict IFN-driven immune dysfunction.

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: Distinct ISGs and epigenetic features from cancer cells predict low CD8+ T cell cytolytic activity.
Fig. 2: Widespread remodeling of the relapsed tumor epigenome is characterized by activated enhancers with ISRE-like motifs.
Fig. 3: Tumors resistant to ICB are characterized by H3K4me1-marked inflammatory memory domains regulated by STAT1 and IRF3.
Fig. 4: OAS1 is controlled by inflammatory memory domains and amplifies IFN-I signaling in ICB-resistant cancer cells.
Fig. 5: Blocking OAS1 and IFN-I signaling in tumor cells restores CD8+ T cell-driven response to ICB.
Fig. 6: Inhibiting tumor IFN-I signaling enhances IFN signaling in immune cells to promote DC–CD8+ T cell interactions.
Fig. 7: Inhibiting tumor IFN-I signaling promotes transition of CD8+ T cell clonotypes toward an effector-like state after anti-PD1 therapy.

Similar content being viewed by others

Data availability

Raw sequencing reads and processed data for Figs. 24 and Extended Data Figs. 35 (ATAC-seq, RNA-seq, CUT&RUN) are deposited in the Gene Expression Omnibus under accession number GSE219179. Raw sequencing reads for Figs. 6 and 7 and Extended Data Figs. 7 and 8 (scRNA-seq and scTCR-seq) are deposited in the Sequence Read Archive and available under accession number PRJNA626462. RNA-seq data related to Fig. 5f have been previously deposited in the Gene Expression Omnibus under accession number GSE83850. Human RNA-seq and ATAC-seq tumor data used for Fig. 1 were derived from the TCGA Research Network and downloaded from the UCSC Xena Browser (TCGA PANCAN cohort) and the NCI Genomic Data Commons (https://gdc.cancer.gov/about-data/publications/ATACseq-AWG), respectively. Source data are provided with this paper.

Code availability

Processing and analysis code related to the TCGA and mouse integrated epigenomic analyses is deposited in a GitHub repository at https://github.com/jingyaqiu/ifnar_epigenome.

References

  1. Dighe, A. S., Richards, E., Old, L. J. & Schreiber, R. D. Enhanced in vivo growth and resistance to rejection of tumor cells expressing dominant negative IFNγ receptors. Immunity 1, 447–456 (1994).

  2. Diamond, M. S. et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J. Exp. Med. 208, 1989–2003 (2011).

    Article  CAS  Google Scholar 

  3. Fuertes, M. B. et al. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8α+ dendritic cells. J. Exp. Med. 208, 2005–2016 (2011).

    Article  CAS  Google Scholar 

  4. Teijaro, J. R. et al. Persistent LCMV infection is controlled by blockade of type I interferon signaling. Science 340, 207–211 (2013).

    Article  CAS  Google Scholar 

  5. Wilson, E. B. et al. Blockade of chronic type I interferon signaling to control persistent LCMV infection. Science 340, 202–207 (2013).

    Article  CAS  Google Scholar 

  6. Benci, J. L. et al. Tumor interferon signaling regulates a multigenic resistance program to immune checkpoint blockade. Cell 167, 1540–1554 (2016).

    Article  CAS  Google Scholar 

  7. Benci, J. L. et al. Opposing functions of interferon coordinate adaptive and innate immune responses to cancer immune checkpoint blockade. Cell 178, 933–948 (2019).

    Article  CAS  Google Scholar 

  8. Chen, J. et al. Type I IFN protects cancer cells from CD8+ T cell-mediated cytotoxicity after radiation. J. Clin. Invest. 129, 4224–4238 (2019).

    Article  Google Scholar 

  9. Jacquelot, N. et al. Sustained type I interferon signaling as a mechanism of resistance to PD-1 blockade. Cell Res. 29, 846–861 (2019).

    Article  CAS  Google Scholar 

  10. Dubrot, J. et al. In vivo CRISPR screens reveal the landscape of immune evasion pathways across cancer. Nat. Immunol. 23, 1495–1506 (2022).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  12. Gao, J. et al. Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 167, 397–404 (2016).

    Article  CAS  Google Scholar 

  13. Qiao, Y. et al. Synergistic activation of inflammatory cytokine genes by interferon-γ-induced chromatin remodeling and Toll-like receptor signaling. Immunity 39, 454–469 (2013).

    Article  CAS  Google Scholar 

  14. Park, S. H. et al. Type I interferons and the cytokine TNF cooperatively reprogram the macrophage epigenome to promote inflammatory activation. Nat. Immunol. 18, 1104–1116 (2017).

    Article  CAS  Google Scholar 

  15. Naik, S. et al. Inflammatory memory sensitizes skin epithelial stem cells to tissue damage. Nature 550, 475–480 (2017).

    Article  CAS  Google Scholar 

  16. Twyman-Saint Victor, C. et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 520, 373–377 (2015).

  17. Miller, B. C. et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 20, 326–336 (2019).

    Article  CAS  Google Scholar 

  18. Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016).

    Article  CAS  Google Scholar 

  19. Paley, M. A. et al. Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science 338, 1220–1225 (2012).

    Article  CAS  Google Scholar 

  20. Pauken, K. E. et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 2807, 1160–1165 (2016).

    Article  Google Scholar 

  21. Ghoneim, H. E. et al. De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell 170, 142–157 (2017).

    Article  CAS  Google Scholar 

  22. Wu, T. et al. The TCF1–Bcl6 axis counteracts type I interferon to repress exhaustion and maintain T cell stemness. Sci. Immunol. 1, eaai8593 (2016).

    Article  Google Scholar 

  23. Rooney, M. S., Shukla, S. A., Wu, C. J., Getz, G. & Hacohen, N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 160, 48–61 (2015).

    Article  CAS  Google Scholar 

  24. Corces, M. R. et al. The chromatin accessibility landscape of primary human cancers. Science 362, eaav1898 (2018).

  25. Larsen, S. B. et al. Establishment, maintenance, and recall of inflammatory memory. Cell Stem Cell 28, 1758–1774 (2021).

    Article  CAS  Google Scholar 

  26. Ordovas-Montanes, J., Beyaz, S., Rakoff-Nahoum, S. & Shalek, A. K. Distribution and storage of inflammatory memory in barrier tissues. Nat. Rev. Immunol. 20, 308–320 (2020).

    Article  CAS  Google Scholar 

  27. Malathi, K., Dong, B., Gale, M. & Silverman, R. H. Small self-RNA generated by RNase L amplifies antiviral innate immunity. Nature 448, 816–819 (2007).

    Article  CAS  Google Scholar 

  28. Hornung, V., Hartmann, R., Ablasser, A. & Hopfner, K. P. OAS proteins and cGAS: unifying concepts in sensing and responding to cytosolic nucleic acids. Nat. Rev. Immunol. 14, 521–528 (2014).

    Article  CAS  Google Scholar 

  29. Manguso, R. T. et al. In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target. Nature 547, 413–418 (2017).

    Article  CAS  Google Scholar 

  30. Garris, C. S. et al. Successful anti-PD-1 cancer immunotherapy requires T cell–dendritic cell crosstalk involving the cytokines IFN-γ and IL-12. Immunity 49, 1148–1161 (2018).

    Article  CAS  Google Scholar 

  31. Chow, M. T. et al. Intratumoral activity of the CXCR3 chemokine system is required for the efficacy of anti-PD-1 therapy. Immunity 50, 1498–1512 (2019).

    Article  CAS  Google Scholar 

  32. Zilionis, R. et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50, 1317–1334 (2019).

    Article  CAS  Google Scholar 

  33. Jin, S. et al. Inference and analysis of cell–cell communication using CellChat. Nat. Commun. 12, 1088 (2021).

  34. Tsuyuzaki, K., Ishii, M. & Nikaido, I. Uncovering hypergraphs of cell–cell interaction from single cell RNA-sequencing data. Preprint at bioRxiv https://doi.org/10.1101/566182 (2019).

  35. Zhang, L. et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 564, 268–272 (2018).

    Article  CAS  Google Scholar 

  36. Netea, M. G. et al. Trained immunity: a program of innate immune memory in health and disease. Science 352, aaf1098 (2016).

    Article  Google Scholar 

  37. Ishizuka, J. J. et al. Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade. Nature 565, 43–48 (2019).

    Article  CAS  Google Scholar 

  38. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 2015, 21.29.1–21.29.9 (2015).

    Google Scholar 

  39. Rao, S. S. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).

    Article  CAS  Google Scholar 

  40. Vierstra, J. et al. Global reference mapping of human transcription factor footprints. Nature 583, 729–736 (2020).

    Article  CAS  Google Scholar 

  41. Hudson, W. H. et al. Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1+ stem-like CD8+ T cells during chronic infection. Immunity 51, 1043–1058 (2019).

    Article  CAS  Google Scholar 

  42. Kurtulus, S. et al. Checkpoint blockade immunotherapy induces dynamic changes in PD-1CD8+ tumor-infiltrating T cells. Immunity 50, 181–194 (2019).

    Article  CAS  Google Scholar 

  43. Beltra, J. C. et al. Developmental relationships of four exhausted CD8+ T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity 52, 825–841 (2020).

    Article  CAS  Google Scholar 

  44. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  Google Scholar 

  45. Atchley, W. R., Zhao, J., Fernandes, A. D. & Drüke, T. Solving the protein sequence metric problem. Proc. Natl Acad. Sci. USA 102, 6395–6400 (2005).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

A.J.M., J.Q., S.W., Y.X., E.J.W. and J.S. were supported by the Mark Foundation for Cancer Research. A.J.M., J.Q., B.X., D.Y., J.L.B., J.C.B. and E.J.W. were supported by the Parker Institute for Cancer Immunotherapy. A.J.M., D.Y., H.I. and E.J.W. were also supported by a program project grant from the NIH (1P01CA210944-01), and J.Q. was supported by the National Human Genome Research Institute (5T32HG000046-18). A.J.M. was additionally supported by the Breast Cancer Research Foundation. We thank the Penn Cytomics and Cell Sorting Resource Laboratory staff, staff at the University Laboratory Animal Resources and the Penn Genomics and Sequencing Core staff.

Author information

Authors and Affiliations

Authors

Contributions

J.Q., B.X. and A.J.M. designed experiments. B.X. performed the main experiments; D.R., S.W. and J.S. assisted with epigenome profiling; and J.L.B., Y.X. and D.Y. assisted with mouse and immune profiling studies. J.Q. and A.J.M. designed computational methods. J.Q., B.X. and A.J.M. analyzed data, interpreted results and wrote the manuscript. H.I. assisted with computation methods. J.-C.B. provided data for exhausted T cell subsets. J.-C.B. and E.J.W. assisted in the interpretation of T cell exhaustion studies. A.J.M. oversaw the study.

Corresponding author

Correspondence to Andy J. Minn.

Ethics declarations

Competing interests

A.J.M. is a project member of the Parker Institute for Cancer Immunotherapy and has received research funding from Merck. He has also received honoraria and travel support from Merck, AstraZeneca and Pfizer. He is a scientific founder for Dispatch Biotherapeutics and a scientific consultant for Takeda, Xilio, H3 Biomedicine and Related Sciences. A.J.M. is an inventor on patents related to the IFN pathway and is on a filed patent related to modified CAR T cells. J.L.B. is currently an employee of Bristol Myers Squibb. Y.X. is a current employee of Pfizer. B.X. is currently an employee of Incyte. E.J.W. is a member of the Parker Institute for Cancer Immunotherapy and has consulting agreements with and/or is on the scientific advisory board for Merck, Roche, Pieris, Elstar and Surface Oncology. E.J.W. has a patent licensing agreement on the PD1 pathway with Roche–Genentech and is a founder of Arsenal Biosciences. The remaining authors declare no relevant competing interests.

Peer review

Peer review information

Nature Cancer thanks Zlatko Trajanoski 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 Relationship between ISGs and T cell activity in TCGA tumors.

A. Schematic depicting the datasets and strategy used for the integrated analysis of RNA-seq and ATAC-seq data from TCGA tumor samples. B. CD8 T cell cytolytic activity scores (CD8 cytolytic activity) for all TCGA tumors, ordered by increasing median score (red line) for each cancer type. Cancer types with grey labels were excluded from analysis due to known low immune infiltration or low numbers of samples with paired RNA and ATAC data. C. Standardized regression coefficient estimates representing the effect of modified Z-score normalized RNA expression of IFNG.GS, ISG.RS, or 250 random gene sets (n = 38, same size as ISG.RS gene set) on CD8 T cell cytolytic activity. D. Histogram of number of putative cis-regulatory elements linked to each ISG.RS gene.

Source data

Extended Data Fig. 2 OAS1 cis-regulatory elements in TCGA tumors.

A-C. ATAC tracks at the OAS1 loci for representative LUAD, LUSC, and STAD tumors with high (blue) or low (red) CD8 T cell cytolytic activity. The annotation bar (bottom) demarcates called peaks, with black and grey bars indicating putative cis-REs linked to OAS1 and other peaks in the region, respectively. Highlighted peaks indicate putative cis-REs that negatively correlate with CD8 T cell cytolytic activity. The correlation of average OAS1 cis-RE chromatin accessibility with CD8 T cell cytolytic activity across all paired tumor samples available for each cancer type is depicted in Fig. 1i.

Extended Data Fig. 3 Activated enhancers in ICB-resistant cancer cells.

A. Spearman pairwise correlation of RNA and ATAC libraries. Heatmap is hierarchically clustered. B. PCA of H3K4me3, H3K27ac, and H3K4me1 signals for B16 and Res 499 cells with or without Stat1 knockout (SKO) sorted from in vivo tumors (n = 2 mice per group). C. Strategy for integrated annotation of putative regulatory elements. D. Summary profiles of H3K4me3, H3K27ac, and ATAC signal intensity over activated or deactivated promoters and enhancers (n = 2 mice per condition for H3K4me3 and H3K27ac assays; n = 5 mice per condition for ATAC assay). E. Genomic regions where Res 499 activated or deactivated regulatory elements reside. Only regulatory elements with significant differences in H3K27ac, H3K4me3, or ATAC signal intensity are included (FDR < 0.05). F. Example tracks for activated enhancers in Res 499 that are either pre-existing (left) or de novo activated (right). G. Enriched PANTHER pathways for genes located within a 50 Kb cis-regulatory window of Res 499 activated enhancers. P-values determined by Fisher’s exact test and FDR calculated by the Benjamini-Hochberg procedure. All pathways shown are significant with FDR < 0.01. H. Chromatin accessibility of the subset of Res 499 activated enhancers induced by chronic IFNG signaling (900 out of 3,738 enhancers) in cancer cells sorted from the indicated in vivo tumors (n = 3 mice per condition), where each row represents a chronic IFNG-induced enhancer. Summary enrichment scores for these loci are shown in Fig. 2i. B16 tumors were treated with IFNG in vitro for 6 hours (acute) or 3.5 weeks (chronic) prior to implantation into syngeneic mice. I. Enrichment of archetype motifs in Res 499 activated enhancers that are chronic IFNG-induced or not. P-values are color-coded and larger circle sizes indicate greater significance. TF motifs highlighted in red are associated with IFN signaling, TF motifs highlighted in blue are associated with inflammatory memory. J. Normalized ATAC-seq insertion counts at DNA footprints with ISRE motifs in B16 or Res 499 cells.

Source data

Extended Data Fig. 4 Features of IFN-associated inflammatory memory and resolved domains.

A. Density histogram representing the distribution of H3K27ac, H3K4me1, or ATAC signal intensity at individual Res 499 activated enhancers in cancer cells from B16 or Res 499 WT or Stat1 KO tumors. Dotted line indicates the mean signal intensity of all activated enhancers for the specified condition. P-values determined using the summary enrichment scores for each condition, and a one-way ANOVA with post-hoc Tukey HSD to calculate significance of pairwise comparisons. B. Top GO Biological Processes terms enriched in genes linked to resolved domains. C. Summary enrichment scores of H3K27ac, H3K4me1, or ATAC signal at resolved domains in cancer cells sorted from the indicated in vivo tumors (n = 2 mice per condition for H3K27ac and H3K4me1 assays; n = 5 mice per condition for ATAC assay). D. Representative track for H3K27ac, H3K4me1, and ATAC signal at an IFN-associated inflammatory memory domain (IFN-IMD) (more examples shown in Fig. 3c). Red bars highlight the IFN-IMD domain where persistent memory features specific to Res 499 tumors are revealed by Stat1 KO. E. Summary gene set enrichment scores for the IFN-associated inflammatory memory gene signature from cancer cells sorted from the indicated in vivo tumors. B16 tumors were treated with IFNG in vitro for 6 hours (acute) or 3.5 weeks (chronic) prior to implantation into syngeneic mice (n = 3 mice per condition). P-values determined by two-sided t-test. F. Log2 fold change in RNA expression between Res 499 and B16 tumors for individual genes in the specified gene set. Coral color indicates an inflammatory memory gene that is summarized in Fig. 3f.

Source data

Extended Data Fig. 5 OAS1 regulates IFN-I signaling and IRF3 maintains accessibility of activated enhancers in ICB-resistant cancer cells.

A. Ifnb expression in tumor infiltrating CD45+ immune cells from mice with Res 499 WT or Ifnar1 KO (IFNAR KO) tumors, treated with or without anti-PD1 (n = 29,584 cells from 2 biological replicates per condition). P-values determined by two-sided Wilcoxon test. B. RNA expression of OAS1 in B16 and Res 499 cells in vitro (n = 5 per condition). P-values determined by two-sided t-test. C-D. Protein (C) and RNA (D) expression of OAS1 in TSA and ICB-resistant Res 237 breast cancer cells in vitro (RNA, n = 6 per group). Replicate samples are shown in the protein blot. P-values determined by two-sided t-test. E. Protein expression of OAS1 in Res 499 WT and Oas1 KO cells. Shown is representative of two technical repeats. F. Protein expression of OAS1 following IFNB (1000 U/ml) stimulation in Res 237 control (CTRL) and Res 237 Oas1 KO clones (cl2, 4, 15). Shown is representative of two technical repeats. G. Concentration of IFNA and IFNB after polyI:C transfection of TSA, Res 237 (R237), and Res 237 Oas1 KO clones (cl2, cl4) (TSA, n = 8; Res 237, n = 8; cl2, n = 7; cl4, n = 4 biological replicates). P-values determined by two-sided t-test. H. Oas1 expression in Res 499 control (CTRL) or Res 499 Stat1 KO cells in vitro (left; Res 499 CTRL, n = 10; Res 499 Stat1 KO) or in Res 499 Stat1 KO cells following siRNA knockdown of control RNA (siCTRL) or the indicated IRFs (right; n = 10; siCTRL, n = 10; siIRF1, n = 7; siIRF3, n = 10, siIRF7, n = 7, siIRF9, n = 7). P-values determined by two-sided t-test. I. Chromatin accessibility of individual Res 499 activated enhancers in cancer cells sorted from the indicated in vivo tumors. Boxplots represent the 25th percentile, median, 75th percentile, and 1.5x IQR (whiskers).

Source data

Extended Data Fig. 6 Blocking cancer cell IFN-I signaling increases the frequency of tumor-infiltrating CD8 T cells and improves ICB response.

A. Survival of mice with Res 237 WT or Oas1 KO (KO) tumors either non-treated (NT) or treated with anti-CTLA4 (aCTLA4) (NT, n = 10 mice per condition; aCTLA4, n = 20 mice per condition). Two independent KO clones are shown. P-values determined by two-sided log-rank test. B. Percent of tumor infiltrating CD8 T cells in total CD45+ cells from mice with Res 237 WT or Res 237 Oas1 KO (KO) tumors. P-values determined by two-sided t-test. C. MHC-I surface expression at baseline and after either IFNB or IFNG treatment of the indicated cell lines to assess knockout of B2m and/or Ifnar1 in Res 499 cancer cells. D. Tumor volume growth of mice injected with Res 499 tumors and treated with anti-CTLA4 plus anti-PD1 (dICB), delayed administration of the JAK inhibitor ruxolitinib (JAKi), or both (NT and JAKi, n = 5 mice per condition; dICB and JAKi + dICB, n = 10 mice per condition). P-values determined by a mix-effect regression model. E. Percent of PRF1+ CD8 T cells relative to total CD8 T cells in Res 499 WT or Res 499 Ifnar1 KO (KO) tumors from mice treated with or without anti-PD1 (aPD1) (n = 5 mice per group). P-values determined by two-sided t-test. F-G. Survival of mice with B16 WT or Ifnar1 KO (KO) tumors (F), or CT26 WT or Ifnar1 KO (KO) tumors (G), either non-treated (NT) or treated with anti-PD1 (NT, n = 5 mice per condition; aPD1, n = 10 mice per condition). P-values determined by two-sided log-rank test. H. Representative density plot of peripheral CD45+ cells in control mice or mice treated with anti-CD8 or anti-NK1.1 depleting antibody. I. Data shown in Fig. 5a replotted to emphasize effect of host immune cell status. Boxplots represent the 25th percentile, median, 75th percentile, and 1.5x IQR (whiskers).

Source data

Extended Data Fig. 7 Blocking cancer cell IFN-I signaling improves immune cell IFNG signaling and predicted interactions between DCs and CD8 T cells.

A. Distribution of Ifng and IFNG.GS expression in tumor infiltrating CD45+ immune cells from mice with Res 499 WT or Ifnar1 KO (KO) tumors treated with or without anti-PD1 (aPD1) (n = 29,584 cells from 2 biological replicates per condition). P-values determined by two-sided Wilcoxon test. B. Percent CD8 T cells expressing IFNG by flow cytometry (left; n = 13 mice per group) or average expression of Ifng in CD8 T cell subsets by scRNA-seq (right) from the indicated tumors either non-treated (NT) or treated with anti-PD1. P-values determined by two-sided t-test. For heatmap of Ifng expression, boxed values in second column indicate p < 0.05 for comparison between WT vs. Ifnar1 KO, and boxed values in fourth column are for WT + aPD1 vs. Ifnar1 KO + aPD1. C. Average expression of select markers for indicated DC subtype clusters shown in Fig. 6d. D. Average expression of DC3 ligands and CD8 T cell receptors from the receptor-ligands shown in Fig. 6h. The cell types are annotated in the UMAP on the left. E. Cell-cell interaction scores (left) and ligand-receptor interactions (right) between CD8 T cells and DC3 using scTensor. Data are from mice bearing Res 499 WT or Ifnar1 KO tumors treated with or without anti-PD1. Cell-cell interaction scores and the mean (red dot) are from biological replicates. P-value determined by a one-way ANOVA for differences between groups. F. Surface expression of CD86 on MHC-II+ CD11c+ DCs from Res 499 WT or Ifnar1 KO tumors (n = 9 per condition). P-values determined by two-sided t-test. G. Survival of wildtype (WT) or Batf3−/- mice with Res 499 Ifnar1 KO cells treated with or without anti-PD1 (WT mice NT, n = 5; WT mice aPD1, n = 10; Batf3 -/- mice NT, n = 3; Batf -/- mice aPD1, n = 7). P-values determined by two-sided log-rank test. Boxplots represent the 25th percentile, median, 75th percentile, and 1.5x IQR (whiskers).

Source data

Extended Data Fig. 8 Blocking cancer cell IFN-I signaling alters features of tumor-infiltrating CD8 T cells.

A-B. Expression of CD8 T cell gene sets (A) and average expression of select markers (B) used to annotate CD8 T cell states shown in Fig. 7a. C. Percentage CD8 T cells occupying each state for each biological replicate (n = 2 mice per condition). Black dot represents mean. D. Percent of CX3CR1+ effector-like T cells relative to TIM3- CD8 T cells in Res 499 WT or Res 499 Ifnar1 KO (KO) tumors from mice treated with or without anti-PD1 (aPD1) (n = 10 mice per condition). E-F. UMAP of CD8 T cell clonotypes clustered by biophysical features of TCR CDR3 amino acids (E) and the distribution of expanded T cell clonotypes across TCR clusters (F) in Res 499 WT or Res 499 Ifnar1 KO (KO) tumors from mice treated with or without anti-PD1. In the UMAP, TCR clusters are color-coded and circle size indicates clonotype frequency. In the bar plot, each bar is one unique clonotype stratified by TCR cluster, with the height of the bar representing the reciprocal of the rank order by clonotype frequency (higher values indicate greater clonotype expansion). Boxplots represent the 25th percentile, median, 75th percentile, and 1.5x IQR (whiskers).

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1

Reporting Summary

Supplementary Tables

Supplementary Tables 1–5.

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 Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

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.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qiu, J., Xu, B., Ye, D. et al. Cancer cells resistant to immune checkpoint blockade acquire interferon-associated epigenetic memory to sustain T cell dysfunction. Nat Cancer 4, 43–61 (2023). https://doi.org/10.1038/s43018-022-00490-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-022-00490-y

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