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.

  • Letter
  • Published:

Tumor innate immunity primed by specific interferon-stimulated endogenous retroviruses

Abstract

Mesenchymal tumor subpopulations secrete pro-tumorigenic cytokines and promote treatment resistance1,2,3,4. This phenomenon has been implicated in chemorefractory small cell lung cancer and resistance to targeted therapies5,6,7,8, but remains incompletely defined. Here, we identify a subclass of endogenous retroviruses (ERVs) that engages innate immune signaling in these cells. Stimulated 3 prime antisense retroviral coding sequences (SPARCS) are oriented inversely in 3′ untranslated regions of specific genes enriched for regulation by STAT1 and EZH2. Derepression of these loci results in double-stranded RNA generation following IFN-γ exposure due to bi-directional transcription from the STAT1-activated gene promoter and the 5′ long terminal repeat of the antisense ERV. Engagement of MAVS and STING activates downstream TBK1, IRF3, and STAT1 signaling, sustaining a positive feedback loop. SPARCS induction in human tumors is tightly associated with major histocompatibility complex class 1 expression, mesenchymal markers, and downregulation of chromatin modifying enzymes, including EZH2. Analysis of cell lines with high inducible SPARCS expression reveals strong association with an AXL/MET-positive mesenchymal cell state. While SPARCS-high tumors are immune infiltrated, they also exhibit multiple features of an immune-suppressed microenviroment. Together, these data unveil a subclass of ERVs whose derepression triggers pathologic innate immune signaling in cancer, with important implications for cancer immunotherapy.

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: Discovery of an IFN-inducible subclass of ERVs.
Fig. 2: SPARCS expression is inducible and triggers positive feedback amplification.
Fig. 3: Expression of SPARCS-containing genes across cancers.
Fig. 4: SPARCS-containing gene expression is associated with adaptive and immune-suppressive signatures.

Similar content being viewed by others

References

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

    Article  CAS  Google Scholar 

  2. Marusyk, A. et al. Non-cell-autonomous driving of tumour growth supports sub-clonal heterogeneity. Nature 514, 54–58 (2014).

    Article  CAS  Google Scholar 

  3. Junttila, M. R. & de Sauvage, F. J. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature 501, 346–354 (2013).

    Article  CAS  Google Scholar 

  4. Tabassum, D. P. & Polyak, K. Tumorigenesis: It takes a village. Nat. Rev. Cancer 15, 473–483 (2015).

    Article  CAS  Google Scholar 

  5. Konieczkowski, D. J. et al. A melanoma cell state distinction influences sensitivity to MAPK pathway inhibitors. Cancer Discov. 4, 816–827 (2014).

    Article  CAS  Google Scholar 

  6. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  CAS  Google Scholar 

  7. Wu, X. et al. AXL kinase as a novel target for cancer therapy. Oncotarget 5, 9546–9563 (2014).

    PubMed  PubMed Central  Google Scholar 

  8. Hata, A. N. et al. Tumor cells can follow distinct evolutionary paths to become resistant to epidermal growth factor receptor inhibition. Nat. Med. 22, 262–269 (2016).

    Article  CAS  Google Scholar 

  9. Canadas, I. et al. Targeting epithelial-to-mesenchymal transition with Met inhibitors reverts chemoresistance in small cell lung cancer. Clin. Cancer Res. 20, 938–950 (2014).

    Article  CAS  Google Scholar 

  10. Calbo, J. et al. A functional role for tumor cell heterogeneity in a mouse model of small cell lung cancer. Cancer Cell 19, 244–256 (2011).

    Article  CAS  Google Scholar 

  11. Chiappinelli, K. B. et al. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell 162, 974–986 (2015).

    Article  CAS  Google Scholar 

  12. Roulois, D. et al. DNA-demethylating agents target colorectal cancer cells by inducing viral mimicry by endogenous transcripts. Cell 162, 961–973 (2015).

    Article  CAS  Google Scholar 

  13. Chuong, E. B., Elde, N. C. & Feschotte, C. Regulatory evolution of innate immunity through co-option of endogenous retroviruses. Science 351, 1083–1087 (2016).

    Article  CAS  Google Scholar 

  14. Rouillard, A. D. et al. The harmonizome: A collection of processed datasets gathered to serve and mine knowledge about genes and proteins. Database (Oxford) 2016, baw100 (2016).

    Article  Google Scholar 

  15. Gao, D. et al. Cyclic GMP-AMP synthase is an innate immune sensor of HIV and other retroviruses. Science 341, 903–906 (2013).

    Article  CAS  Google Scholar 

  16. Poirier, J. T. et al. DNA methylation in small cell lung cancer defines distinct disease subtypes and correlates with high expression of EZH2. Oncogene 34, 5869–5878 (2015).

    Article  CAS  Google Scholar 

  17. Henke, C. et al. Selective expression of sense and antisense transcripts of the sushi-ichi-related retrotransposon-derived family during mouse placentogenesis. Retrovirology 12, 9 (2015).

    Article  Google Scholar 

  18. Hoadley, K. A. et al. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell 158, 929–944 (2014).

    Article  CAS  Google Scholar 

  19. Kim, J. W. et al. Characterizing genomic alterations in cancer by complementary functional associations. Nat. Biotechnol. 34, 539–546 (2016).

    Article  CAS  Google Scholar 

  20. Rhodes, D. A., Reith, W. & Trowsdale, J. Regulation of immunity by butyrophilins. Annu. Rev. Immunol. 34, 151–172 (2016).

    Article  CAS  Google Scholar 

  21. Bindea, G. et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 39, 782–795 (2013).

    Article  CAS  Google Scholar 

  22. Jenkins, R. W. et al. Ex vivo profiling of PD-1 blockade using organotypic tumor spheroids. Cancer Discov. 8, 196–215 (2017).

    Article  Google Scholar 

  23. Pan, D. et al. A major chromatin regulator determines resistance of tumor cells to T cell-mediated killing. Science 359, 770–775 (2018).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  25. Peng, D. et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature 527, 249–253 (2015).

    Article  CAS  Google Scholar 

  26. 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 

  27. Christensen, C. L. et al. Targeting transcriptional addictions in small cell lung cancer with a covalent CDK7 inhibitor. Cancer Cell 26, 909–922 (2014).

    Article  CAS  Google Scholar 

  28. Aref, A. R. et al. Screening therapeutic EMT blocking agents in a three-dimensional microenvironment. Integr. Biol. (Camb.) 5, 381–389 (2013).

    Article  CAS  Google Scholar 

  29. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    Article  CAS  Google Scholar 

  30. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    Article  CAS  Google Scholar 

  31. Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    Article  CAS  Google Scholar 

  32. Perocchi, F., Xu, Z., Clauder-Munster, S. & Steinmetz, L. M. Antisense artifacts in transcriptome microarray experiments are resolved by actinomycin D. Nucleic Acids Res. 35, e128 (2007).

    Article  Google Scholar 

  33. Chen, J. et al. Over 20% of human transcripts might form sense-antisense pairs. Nucleic Acids Res. 32, 4812–4820 (2004).

    Article  CAS  Google Scholar 

  34. Carey, C. D. et al. Topological analysis reveals a PD-L1-associated microenvironmental niche for Reed–Sternberg cells in Hodgkin lymphoma. Blood 130, 2420–2430 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  36. Barbie, D. A. et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature 462, 108–112 (2009).

    Article  CAS  Google Scholar 

  37. Barretina, J. et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012).

    Article  CAS  Google Scholar 

  38. Sen, D. R. et al. The epigenetic landscape of T cell exhaustion. Science 354, 1165–1169 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Albanell, A. Rovira, and E. Arriola (Hospital del Mar Medical Research Institute, Barcelona, Spain) for providing human SCLC cell lines and the H69/H69M cell model. We also thank W.G. Kaelin, Jr. (Dana–Farber Cancer Institute, Boston, MA) for providing human ccRCC cell lines. This work was supported by NCI-R01 CA190394-02 and NIH-U01 CA2143A1-01 (D.A.B.), the Gloria T. Maheu, Steven J. Schaubert, and Heerwagen Family Funds for Lung Cancer Research (D.A.B.), the Rising Tide Foundation (D.A.B.), NIH-U01 CA217885 (J.W.K., P.T.), NIH/NCI P01CA120964 (K.K.W.), 5R01CA163896-04 (K.K.W.), 5R01CA140594-07 (K.K.W.), 5R01CA122794-10 (K.K.W.), 5R01CA166480-04 (K.K.W.), the Gross-Loh Family Fund for Lung Cancer Research (K.K.W., D.A.B.), and the Susan Spooner Family Lung Cancer Research Fund at Dana–Farber Cancer Institute (K.K.W.). Additional funding was provided by NIH grants P01 CA114046, P01 CA025874, P30 CA010815, and R01 CA047159 and by the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation and the Melanoma Research Foundation. The support for Shared Resources used in this study was provided by Cancer Center Support Grant CA010815 (to The Wistar Institute). Additional support from a Stand Up To Cancer–American Cancer Society Lung Cancer Dream Team Translational Research Grant (SU2CAACR-DT1715). 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. R.T. is a Howard Hughes Medical Institute Medical Research Fellow.

Author information

Authors and Affiliations

Authors

Contributions

I.C. and D.A.B. designed the research and wrote the manuscript. J.W.K., G.Z., T.Ti., D.M., P.T., Z.W., M.H., and H.W. performed and supervised computational analyses. I.C., R.T., S.Ki., R.W.J., M.C., T.Th., B.P., H.T., A.R.A., S.Ko., T.U.B., R.U., K.K.W., and D.A.B. performed and supervised biological and cellular studies. C.L.C., Y.I., T.H., M.W., H.B., A.E.A., C.A.L., L.M.S., E.S., and J.S. obtained samples and performed or supervised immunohistochemistry. E.G. and S.R. performed and supervised multiplexed immunofluorescence. D.R.S. and H.W. performed ATAC sequencing and analysis. C.L.C. and Y.Z. performed in vivo experiments. C.P.P. and Y.K. performed ddPCR experiments.

Corresponding author

Correspondence to David Allen Barbie.

Ethics declarations

Competing interests

D.A.B. is a consultant for N-of-One.

Additional information

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

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15 and Supplementary Table 1

Reporting Summary

Supplementary Dataset 1

List of 3(UTR repeat elements from RefSeq and list of 452 top genes upregulated/downregulated in H69M versus H69

Supplementary Dataset 2

List of gene sets and genes coexpressed with the SPARCS signature across cancers in the TCGA and CCLE datasets

Supplementary Dataset 3

List of gene sets coexpressed with control 3(UTR antisense ERVs from H69M downregulated genes across the TCGA dataset

Supplementary Dataset 4

List of chromosomal alterations associated with the SPARCS signature across the TCGA and CCLE datasets

Supplementary Dataset 5

List of the top 200 genes that overlap after intersecting the top 1,000 genes co-regulated with SPARCS from the TCGA and CCLE datasets

Supplementary Dataset 6

List of cell lines from CCLE or tumors from TCGA ranked based on SPARCS score and RPKM gene expression values of indicated genes

Supplementary Dataset 7

Sequences used in this study

Supplementary Dataset 8

List of genes tested in the OncoPanel Assay

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cañadas, I., Thummalapalli, R., Kim, J.W. et al. Tumor innate immunity primed by specific interferon-stimulated endogenous retroviruses. Nat Med 24, 1143–1150 (2018). https://doi.org/10.1038/s41591-018-0116-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0116-5

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