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:

Autophagy promotes growth of tumors with high mutational burden by inhibiting a T-cell immune response

A Publisher Correction to this article was published on 16 August 2021

This article has been updated

Abstract

Macroautophagy (hereafter autophagy) degrades and recycles intracellular components to sustain metabolism and survival during starvation. Host autophagy promotes tumor growth by providing essential tumor nutrients. Autophagy also regulates immune cell homeostasis and function and suppresses inflammation. Although host autophagy does not promote a T-cell antitumor immune response in tumors with low tumor mutational burden (TMB), whether this was the case in tumors with high TMB was not known. Here we show that autophagy, especially in the liver, promotes tumor immune tolerance by enabling regulatory T-cell function and limiting stimulator of interferon genes, T-cell response and interferon-γ, which enables growth of high-TMB tumors. We have designated this as hepatic autophagy immune tolerance. Autophagy thereby promotes tumor growth through both metabolic and immune mechanisms depending on mutational load and autophagy inhibition is an effective means to promote an antitumor T-cell response in high-TMB tumors.

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: Loss of host autophagy increases the level of circulating pro-inflammatory cytokines and promotes T-cell infiltration in high-TMB tumors.
Fig. 2: Host autophagy promotes high-TMB tumor growth through a T-cell-dependent mechanism.
Fig. 3: Loss of autophagy upregulates an antitumor immune response in a T-cell-dependent manner and increases Treg cells.
Fig. 4: Host autophagy promotes high-TMB tumor growth through suppression of Sting.
Fig. 5: Loss of autophagy increases immune cell fraction and IFN-γ pathway gene expression.
Fig. 6: Loss of autophagy decreases tumor growth in an IFN-γ- and MHC-I-dependent manner.
Fig. 7: Liver autophagy is necessary to maintain tumor growth in an IFN-γ-dependent manner.
Fig. 8: Model depicting hepatic autophagic immune tolerance.

Similar content being viewed by others

Data availability

10X scRNA-seq data have been deposited in the Gene Expression Omnibus under accession ID GSE154654. Whole-exome sequencing data generated in this study can be found in the Gene Expression Omnibus under accession ID GSE155556. All other data supporting the findings of this study are available from the corresponding author upon reasonable request. Source data are provided with this paper.

Change history

References

  1. Karsli-Uzunbas, G. et al. Autophagy is required for glucose homeostasis and lung tumor maintenance. Cancer Discov. 4, 914–927 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Komatsu, M. et al. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J. Cell Biol. 169, 425–434 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Kuma, A. et al. The role of autophagy during the early neonatal starvation period. Nature 432, 1032–1036 (2004).

    CAS  PubMed  Google Scholar 

  4. Guo, J. Y. et al. Autophagy provides metabolic substrates to maintain energy charge and nucleotide pools in Ras-driven lung cancer cells. Genes Dev. 30, 1704–1717 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Kamada, Y., Sekito, T. & Ohsumi, Y. Autophagy in yeast: a TOR-mediated response to nutrient starvation. Curr. Top. Microbiol. Immunol. 279, 73–84 (2004).

    CAS  PubMed  Google Scholar 

  6. Mizushima, N., Levine, B., Cuervo, A. M. & Klionsky, D. J. Autophagy fights disease through cellular self-digestion. Nature 451, 1069–1075 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Kimmelman, A. C. & White, E. Autophagy and tumor metabolism. Cell Metab. 25, 1037–1043 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Amaravadi, R., Kimmelman, A. C. & White, E. Recent insights into the function of autophagy in cancer. Genes Dev. 30, 1913–1930 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Poillet-Perez, L. & White, E. Role of tumor and host autophagy in cancer metabolism. Genes Dev. 33, 610–619 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Sousa, C. M. et al. Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature 536, 479–483 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Yang, A. et al. Autophagy sustains pancreatic cancer growth through both cell-autonomous and nonautonomous mechanisms. Cancer Discov. 8, 276–287 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Poillet-Perez, L. et al. Autophagy maintains tumour growth through circulating arginine. Nature 563, 569–573 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Degenhardt, K. et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 10, 51–64 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Mathew, R. et al. Functional role of autophagy-mediated proteome remodeling in cell survival signaling and innate immunity. Mol. Cell 55, 916–930 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Merkley, S. D., Chock, C. J., Yang, X. O., Harris, J., Castillo, E. F. & Modulating, T. Cell responses via autophagy: the intrinsic influence controlling the function of both antigen-presenting cells and T cells. Front. Immunol. 9, 2914 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. McFadden, D. G. et al. Mutational landscape of EGFR-, MYC-, and Kras-driven genetically engineered mouse models of lung adenocarcinoma. Proc. Natl Acad. Sci. USA 113, E6409–E6417 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Bignell, G. R. et al. Signatures of mutation and selection in the cancer genome. Nature 463, 893–898 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Westcott, P. M. et al. The mutational landscapes of genetic and chemical models of Kras-driven lung cancer. Nature 517, 489–492 (2015).

    CAS  PubMed  Google Scholar 

  19. Wang, J. et al. UV-induced somatic mutations elicit a functional T cell response in the YUMMER1.7 mouse melanoma model. Pigment Cell Melanoma Res. 30, 428–435 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Wei, S. C., Duffy, C. R. & Allison, J. P. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 8, 1069–1086 (2018).

    PubMed  Google Scholar 

  21. Guo, J. Y. et al. Autophagy suppresses progression of K-Ras-induced lung tumors to oncocytomas and maintains lipid homeostasis. Genes Dev. 27, 1447–1461 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Wei, H. et al. Suppression of autophagy by FIP200 deletion inhibits mammary tumorigenesis. Genes Dev. 25, 1510–1527 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Schumacher, T. N. & Schreiber, R. D. Neoantigens in cancer immunotherapy. Science 348, 69–74 (2015).

    CAS  PubMed  Google Scholar 

  24. Panda, A. et al. Identifying a clinically applicable mutational burden threshold as a potential biomarker of response to immune checkpoint therapy in solid tumors. JCO Precis. Oncol. https://doi.org/10.1200/PO.17.00146 (2017).

  25. Yang, A. S., Monken, C. E. & Lattime, E. C. Intratumoral vaccination with vaccinia-expressed tumor antigen and granulocyte macrophage colony-stimulating factor overcomes immunological ignorance to tumor antigen. Cancer Res. 63, 6956–6961 (2003).

    CAS  PubMed  Google Scholar 

  26. Poillet-Perez, L. et al. Autophagy maintains tumour growth through circulating arginine. Nature 563, 569–573 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Chu, H. et al. Gene-microbiota interactions contribute to the pathogenesis of inflammatory bowel disease. Science 352, 1116–1120 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Wei, J. et al. Autophagy enforces functional integrity of regulatory T cells by coupling environmental cues and metabolic homeostasis. Nat. Immunol. 17, 277–285 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Sawant, D. V. et al. Adaptive plasticity of IL-10(+) and IL-35(+) Treg cells cooperatively promotes tumor T cell exhaustion. Nat. Immunol. 20, 724–735 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Sliter, D. A. et al. Parkin and PINK1 mitigate STING-induced inflammation. Nature 561, 258–262 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Castro, F., Cardoso, A. P., Goncalves, R. M., Serre, K. & Oliveira, M. J. Interferon-γ at the crossroads of tumor immune surveillance or evasion. Front. Immunol. 9, 847 (2018).

    PubMed  PubMed Central  Google Scholar 

  32. Yamamoto, K. et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature 581, 100–105 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Cunha, L. D. et al. LC3-associated phagocytosis in myeloid cells promotes tumor immune tolerance. Cell 175, 429–441 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Lee, H. K. et al. In vivo requirement for Atg5 in antigen presentation by dendritic cells. Immunity 32, 227–239 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Xu, X. et al. Autophagy is essential for effector CD8(+) T cell survival and memory formation. Nat. Immunol. 15, 1152–1161 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. DeVorkin, L. et al. Autophagy regulation of metabolism is required for CD8(+) T cell anti-tumor immunity. Cell Rep. 27, 502–513 (2019).

    CAS  PubMed  Google Scholar 

  37. Yu, Y. et al. STING-mediated inflammation in Kupffer cells contributes to progression of nonalcoholic steatohepatitis. J. Clin. Invest. 129, 546–555 (2019).

    PubMed  Google Scholar 

  38. Appleman, L. J. et al. Targeting autophagy and immunotherapy with hydroxychloroquine and interleukin 2 in patients with metastatic renal cell carcinoma (mRCC): a Cytokine Working Group study. J. Clin. Oncol. 36, 106–106 (2018).

    Google Scholar 

  39. Hara, T. et al. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 441, 885–889 (2006).

    CAS  PubMed  Google Scholar 

  40. Gan, B. et al. Role of FIP200 in cardiac and liver development and its regulation of TNF-α and TSC-mTOR signaling pathways. J. Cell Biol. 175, 121–133 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Meeth, K., Wang, J. X., Micevic, G., Damsky, W. & Bosenberg, M. W. The YUMM lines: a series of congenic mouse melanoma cell lines with defined genetic alterations. Pigment Cell Melanoma Res. 29, 590–597 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Summerhayes, I. C. & Franks, L. M. Effects of donor age on neoplastic transformation of adult mouse bladder epithelium in vitro. J. Natl Cancer Inst. 62, 1017–1023 (1979).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Cibulskis, K. et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 31, 213–219 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by National Institutes of Health grants: R01CA193970 (to E.W.), R01CA163591 (to E.W. and J.D.R.), R01CA193970 (to J.M.M.), R01CA211066 (to J.L.G), R01CA243547 (to E.W., S.G. and E.C.L.) and P30CA072720 (Rutgers Cancer Institute of New Jersey) and by the Krauss Foundation (to E.W. and E.C.L.). C.S.C. is supported in part by Stand Up to Cancer and National Science Foundation grant no. 1546101. L.P.P. and S.V.L. received support from a postdoctoral fellowship from the New Jersey Commission for Cancer Research (DHFS16PPC034 and DCHS19PPC009, respectively). Services, results and/or products in support of the research project were generated by funding from the Rutgers Cancer Institute of New Jersey Comprehensive Genomics and Immune Monitoring Shared Resources supported, in part by NCI-CCSG P30CA072720-5920 and by the Lewis-Sigler Institute for Integrative Genomics (Genomics Core Facility).

Author information

Authors and Affiliations

Authors

Contributions

L.P.P. performed the majority of the experimental work and wrote the manuscript. D.W.S. performed/assisted with the MB49 allografts and CD4/CD8 or CD25 depletion and NanoString analysis. Y.Y. developed the methods and provided the mice required for generating Atg5∆/∆ hosts. S.V.L. and C.S.C. analyzed the whole-exome sequencing data and scRNA-seq data. M.I. assisted with the in vivo experiments. P.K.B. assisted with tumor processing and analyzed the data for the NanoString. Z.S.H. assisted with animal husbandry. J.V. performed the flow cytometry, analyzed T-cell exhaustion markers and performed cell isolation and library preparation for single-cell sequencing. M.W.B. provided the YUMM1.1 melanoma cells. J.D.R and S.G provided advice throughout the project and on the manuscript. J.C. provided expertise on CRISPR Cas9. M.H and J.L.G provided the Fip200flox/flox mice. J.M.M. provided expertise in melanoma. E.W. and E.C.L. are the leading principal investigators who conceived the project, supervised research and edited the paper.

Corresponding author

Correspondence to Eileen White.

Ethics declarations

Competing interests

E.W. is co-founder of Vescor Therapeutics and is a consultant for Novartis. S.G. has consulted for Roche, Merck, Novartis, Foundation Medicine and Foghorn Therapeutics. The other authors declare no competing interests.

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 Loss of host autophagy does not decrease the growth of low TMB tumors in a T cell-dependent manner.

a, CD3+, CD4+, CD8+ T cells (percentage) in high TMB tumors (MB49) grown on female Atg7+/+ and Atg7Δ/Δ (n = 5 mice each) hosts following αCD4/8, analyzed by flow cytometry. b, Representative IHC images and quantification of CD3+, CD4+ and CD8+ T cells in YUMM1.3 tumors from male Atg7+/+ and Atg7Δ/Δ hosts (n = 5 mice each). c, d, e, f, Comparison of YUMM1.3 tumor growth on male (c, d) Atg7+/+, Atg7+/+ + αCD4/8, Atg7Δ/Δ and Atg7Δ/Δ + αCD4/8 (n = 10 tumors each) and female (e, f) Atg7+/+(n = 6 tumors), Atg7+/+ + αCD4/8 (n = 12 tumors), Atg7Δ/Δ (n = 12 tumors) and Atg7Δ/Δ + αCD4/8 (n = 12 tumors). Tumor pictures (c, e), and tumor weight and ratio (antibody treated/control) (d, f) from the experimental endpoint. g, h, Comparison of MB49 tumor growth on female Atg7+/+(n = 14 tumors), Atg7+/+ + αCD4 (n = 12 tumors), Atg7+/+ + αCD8 (n = 12 tumors), Atg7+/+ + αCD4/8 (n = 8 tumors), Atg7Δ/Δ (n = 12 tumors), Atg7Δ/Δ + αCD4 (n = 14 tumors), Atg7Δ/Δ + αCD8 (n = 14 tumors) and Atg7Δ/Δ + αCD4/8 (n = 12 tumors). Tumor pictures (g), and tumor weight and ratio (antibody treated/control) (h) from the experimental endpoint. All data are mean +/- s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.001 using two-sided Student’s t-test. Source data available for a, b, d, f, h.

Source data

Extended Data Fig. 2 Loss of host autophagy through Atg5 deletion decreases tumor growth through a T cell-dependent mechanism.

a, Experimental design to induce conditional whole-body Atg5 deletion (Atg5Δ/Δ), and wild-type (Atg5+/+) controls without and with T cell depletion. Ubc-CreERT2/+;Atg5+/+ and Ubc-CreERT2/+;Atg5flox/flox mice were injected with TAM to delete Atg5 and were then injected subcutaneously with tumor cells and intraperitoneally with αCD4/8. Tumor growth was monitored over three weeks. b, c, d, e, Comparison of MB49 tumor growth on male (b, c) Atg5+/+(n = 8 tumors), Atg5+/+ + αCD4/8 (n = 6 tumors), Atg5Δ/Δ (n = 8 tumors) and Atg5Δ/Δ + αCD4/8 (n = 8 tumors) and female (d, e) Atg5+/+(n = 12 tumors), Atg5+/+ + αCD4/8 (n = 10 tumors), Atg5Δ/Δ (n = 12 tumors) and Atg5Δ/Δ + αCD4/8 (n = 10 tumors). Tumor pictures (b, d), and tumor weight and ratio (antibody treated/control) (c, e) from the experimental endpoint. f, Representative IHC images and quantification of CD3+, CD4+ and CD8+ T cells in MB49 tumors from male Atg5+/+ and Atg5Δ/Δ hosts (n = 5 mice each). g, h, Comparison of UV YUMM1.1-9 tumor growth on male (g) Atg7+/+ (n = 10 tumors) and Atg7Δ/Δ (n = 8 tumors) and female (h) Atg7+/+ (n = 16 tumors) and Atg7Δ/Δ (n = 12 tumors) hosts. Tumor pictures and tumor weight from the experimental endpoint. All data are mean +/- s.e.m. *P < 0.05, ***P < 0.001, ****P < 0.0001 using two-sided Student’s t-test. Source data available for c, e, f, g, h.

Source data

Extended Data Fig. 3 Loss of autophagy reduces Tregs and increases immune cell fraction.

a, Overall NanoString gene expression analysis (750 genes) from tumors on female Atg7+/+ and Atg7Δ/Δ hosts with or without αCD4/8 (n = 2 tumors each). Red: high, blue: low expression level. b, Representative IHC images and quantification of FOXP3+ cells in MB49 tumors from female Atg7+/+ and Atg7Δ/Δ hosts (n = 5 tumors each). c, FOXP3+ Treg cells, CD4+ and CD8+ T cells (percentage) in high TMB tumors (MB49) grown on female (n = 3 tumors) Atg7+/+ and Atg7Δ/Δ hosts following αCD25, analyzed by flow cytometry. d, Representative IHC images and quantification of FOXP3+ cells in MB49 tumors from female Atg7+/+ and Atg7Δ/Δ hosts following αCD25 (n = 3 tumors each). e, Survival analysis of Atg7+/+ (n = 19 mice), Atg7Δ/Δ (n = 19 mice), Stinggt/gt;Atg7+/+ (n = 15 mice) and Stinggt/gt;Atg7Δ/Δ (n = 13 mice) mice. f, scRNA-seq UMAP projection of cell cluster from tumors from female Atg7+/+ and Atg7Δ/Δ hosts (n = 3 tumors each). All data are mean +/- s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 using two-sided Student’s t-test. Source data available for b, c, d.

Source data

Extended Data Fig. 4 Loss of autophagy increases high TMB tumor growth in an IFNγ-dependent manner.

a, Survival analysis of Atg7+/+ (n = 16 mice), Atg7Δ/Δ (n = 14 mice), IFNγ-/-;Atg7+/+ (n = 6 mice) and IFNγ-/-;Atg7Δ/Δ (n = 5 mice) hosts. b, Representative H&E tissue staining from IFNγ-/-;Atg7+/+ and IFNγ-/-;Atg7Δ/Δ hosts. Images are representative of two independent experiments. c, d, e, f. Comparison of UV YUMM1.1-9 tumor growth on male (c, d) Atg7+/+ (n = 18 tumors), IFNγ-/-;Atg7+/+ (n = 18 tumors), Atg7Δ/Δ (n = 10 tumors), IFNγ-/-;Atg7Δ/Δ (n = 18 tumors) and female (e, f) Atg7+/+ (n = 16 tumors), IFNγ-/-;Atg7+/+ (n = 18 tumors), Atg7Δ/Δ (n = 16 tumors), IFNγ-/-;Atg7Δ/Δ (n = 18 tumors) hosts. Tumor pictures (c, e), and tumor weight and ratio (antibody treated/control) (d, f) from the experimental endpoint. g, Cropped Western blotting showing expression of B2m in MB49 shC and shB2m tumors from Atg7+/+ and Atg7Δ/Δ hosts (n = 3 tumors each) from one experiment. Actin was used a loading control. All data are mean +/- s.e.m. **P < 0.01, ***P < 0.001 using two-sided Student’s t-test. Source data available for d, f.

Source data

Extended Data Fig. 5 Loss of autophagy, not LC3-associated phagocytosis (LAP), decreases tumor growth through a T cell-dependent mechanism.

a, Serum and b, liver cytokine and chemokine profiling of female and male hosts bearing MB49 tumors with or without liver-specific deletion of Atg7 (n = 6 mice each), showing those with significant differences among 26. c, Experimental design to induce liver-specific Fip200 deletion (liver Fip200Δ/Δ) and wild-type controls (liver Fip200+/+). Fip200flox/flox or C57BL/6J mice were injected with AAV-TBG-iCre and MB49 cells were then injected subcutaneously and tumor growth was monitored over three weeks. d, Cropped Western blotting showing expression of Fip200 in livers, kidneys and brains (n = 3 each) from one experiment. e, Comparison of MB49 tumor growth on male liver Fip200+/+(n = 8 tumors) and liver Fip200Δ/Δ (n = 6 tumors) hosts. f, Experimental design to induce conditional Fip200 deletion (Fip200Δ/Δ) and wild-type (Fip200+/+) controls. C57BL/6J mice were injected with lentiCRISPR scramble or Fip200 to delete Fip200 and were then injected subcutaneously with tumor cells. Tumor growth was monitored over three weeks. g, Representative surveyor assay for Fip200 gRNA screening using liver gDNA from Fip200+/+ and Fip200Δ/Δ hosts (n = 10 mice each) from one experiment. h, Representative IHC images of FIP200+ and p62+ cells in liver from Fip200+/+ and Fip200Δ/Δ hosts (n = 10 mice each). i, j, Comparison of MB49 tumor growth on female Fip200+/+ and Fip200Δ/Δ hosts with or without CD4/8 depletion (n = 10 tumors each). Tumor pictures (i), and tumor weight and ratio (antibody treated/control) (j) from the experimental endpoint. Data are mean +/- s.e.m. *P < 0.05, **P < 0.01, ***P<0.001, ****P < 0.0001 using two-sided Student’s t-test. Source data available for a, b, e, j.

Source data

Supplementary information

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. 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. 2

Statistical Source Data

Source Data Extended Data Fig. 3

Statistical Source Data

Source Data Extended Data Fig. 4

Statistical Source Data

Source Data Extended Data Fig. 5

Statistical Source Data

Source Extended Data Fig. 4

Unprocessed western blots and gels

Source Extended Data Fig. 5

Unprocessed western blots and gels

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Poillet-Perez, L., Sharp, D.W., Yang, Y. et al. Autophagy promotes growth of tumors with high mutational burden by inhibiting a T-cell immune response. Nat Cancer 1, 923–934 (2020). https://doi.org/10.1038/s43018-020-00110-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-020-00110-7

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer