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:

Inhibiting PD-L1 palmitoylation enhances T-cell immune responses against tumours

An Author Correction to this article was published on 11 April 2019

This article has been updated

Abstract

Checkpoint blockade therapy targeting the programmed-death ligand 1 (PD-L1) and its receptor programmed cell death 1 promotes T-cell-mediated immunosurveillance against tumours, and has been associated with marked clinical benefit in cancer patients. Antibodies against PD-L1 function by blocking PD-L1 on the cell surface, but intracellular storage of PD-L1 and its active redistribution to the cell membrane can minimize the therapeutic benefits, which highlights the importance of targeting PD-L1 throughout the whole cell. Here, we show that PD-L1 is palmitoylated in its cytoplasmic domain, and that this lipid modification stabilizes PD-L1 by blocking its ubiquitination, consequently suppressing PD-L1 degradation by lysosomes. We identified palmitoyltransferase ZDHHC3 (DHHC3) as the main acyltransferase required for the palmitoylation of PD-L1, and show that the inhibition of PD-L1 palmitoylation via 2-bromopalmitate, or the silencing of DHHC3, activates antitumour immunity in vitro and in mice bearing MC38 tumour cells. We also designed a competitive inhibitor of PD-L1 palmitoylation that decreases PD-L1 expression in tumour cells to enhance T-cell immunity against the tumours. These findings suggest new strategies for overcoming PD-L1-mediated immune evasion in cancer.

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: Targeting palmitoylation-depleted PD-L1 in tumour cells.
Fig. 2: Identification of the palmitoylation site on PD-L1.
Fig. 3: An intrinsic lysosomal sorting signal in PD-L1 is masked by palmitoylation.
Fig. 4: PD-L1 is palmitoylated by DHHC3 acyltransferase.
Fig. 5: Palmitoylation blocks PD-L1 ubiquitination and ESCRT-mediated trafficking to the MVB.
Fig. 6: Targeting PD-L1 palmitoylation-activated anti-cancer immunity.
Fig. 7: Targeting PD-L1 palmitoylation with a peptidic inhibitor.
Fig. 8: Palmitoylation stabilizes PD-L1 by blocking its ubiquitination and lysosomal degradation.

Similar content being viewed by others

Data availability

The authors declare that all data supporting the findings of this study are available within the paper and its Supplementary Information files.

Change history

  • 11 April 2019

    In the version of this Article originally published, ‘palmitoyltransferase ZDHHC3 (DHHC3)’ was incorrectly referred to as an ‘acetyltransferase’ rather than an as an ‘acyltransferase’; this has now been corrected in five instances. In Fig. 3a, the label for the bottom row of the blots was mistakenly written as ‘GAPHD’; it should have read ‘GAPDH’. In the two right-most panels of Fig. 4j, the antibody labels ‘α-PD-L1’ for the reciprocal co-immunoprecipitation of DHHC3 were incorrect; they should have been ‘α-DHHC3’. These errors have been corrected in all versions of the Article.

References

  1. Brahmer, J. R. et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 366, 2455–2465 (2012).

    Article  CAS  Google Scholar 

  2. Sonpavde, G. PD-1 and PD-L1 inhibitors as salvage therapy for urothelial carcinoma. N. Engl. J. Med. 376, 1073–1074 (2017).

    Article  Google Scholar 

  3. Yao, H., Wang, H., Li, C., Fang, J. Y. & Xu, J. Cancer cell-intrinsic PD-1 and implications in combinatorial immunotherapy. Front. Immunol. 9, 1774 (2018).

    Article  Google Scholar 

  4. Rodell, C. B. et al. TLR7/8-agonist-loaded nanoparticles promote the polarization of tumour-associated macrophages to enhance cancer immunotherapy. Nat. Biomed. Eng. 2, 578–588 (2018).

    Article  Google Scholar 

  5. Sharpe, A. H. & Pauken, K. E. The diverse functions of the PD1 inhibitory pathway. Nat. Rev. Immunol. 18, 153–167 (2018).

    Article  CAS  Google Scholar 

  6. Burr, M. L. et al. CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity. Nature 549, 101–105 (2017).

    Article  CAS  Google Scholar 

  7. Zerdes, I., Matikas, A., Bergh, J., Rassidakis, G. Z. & Foukakis, T. Genetic, transcriptional and post-translational regulation of the programmed death protein ligand 1 in cancer: biology and clinical correlations. Oncogene 37, 4639–4661 (2018).

    Article  CAS  Google Scholar 

  8. Maj, T. et al. Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor. Nat. Immunol. 18, 1332–1341 (2017).

    Article  CAS  Google Scholar 

  9. Snyder, A. et al. Contribution of systemic and somatic factors to clinical response and resistance to PD-L1 blockade in urothelial cancer: an exploratory multi-omic analysis. PLoS Med. 14, e1002309 (2017).

    Article  Google Scholar 

  10. Takeda, Y. et al. A TLR3-specific adjuvant relieves innate resistance to PD-L1 blockade without cytokine toxicity in tumor vaccine immunotherapy. Cell Rep. 19, 1874–1887 (2017).

    Article  CAS  Google Scholar 

  11. Tang, H. et al. Facilitating T cell infiltration in tumor microenvironment overcomes resistance to PD-L1 blockade. Cancer Cell 29, 285–296 (2016).

    Article  CAS  Google Scholar 

  12. Bellucci, R. et al. Interferon-gamma-induced activation of JAK1 and JAK2 suppresses tumor cell susceptibility to NK cells through upregulation of PD-L1 expression. OncoImmunology 4, e1008824 (2015).

    Article  Google Scholar 

  13. Wolfle, S. J. et al. PD-L1 expression on tolerogenic APCs is controlled by STAT-3. Eur. J. Immunol. 41, 413–424 (2011).

    Article  Google Scholar 

  14. Casey, S. C. et al. MYC regulates the antitumor immune response through CD47 and PD-L1. Science 352, 227–231 (2016).

    Article  CAS  Google Scholar 

  15. Bi, X. W. et al. PD-L1 is upregulated by EBV-driven LMP1 through NF-κB pathway and correlates with poor prognosis in natural killer/T-cell lymphoma. J. Hematol. Oncol. 9, 109 (2016).

    Article  Google Scholar 

  16. Lim, S. O. et al. Deubiquitination and stabilization of PD-L1 by CSN5. Cancer Cell 30, 925–939 (2016).

    Article  CAS  Google Scholar 

  17. Mognol, G. P. et al. Exhaustion-associated regulatory regions in CD8+ tumor-infiltrating T cells. Proc. Natl Acad. Sci. USA 114, E2776–E2785 (2017).

    Article  CAS  Google Scholar 

  18. Wang, Y. et al. Regulation of PD-L1: emerging routes for targeting tumor immune evasion. Front. Pharmacol. 9, 536 (2018).

    Article  Google Scholar 

  19. Zhang, J. et al. Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Nature 553, 91–95 (2018).

    Article  CAS  Google Scholar 

  20. Li, C. W. et al. Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity. Nat. Commun. 7, 12632 (2016).

    Article  CAS  Google Scholar 

  21. Wang, H. et al. PD-L2 expression in colorectal cancer: independent prognostic effect and targetability by deglycosylation. OncoImmunology 6, e1327494 (2017).

    Article  Google Scholar 

  22. Li, C. W. et al. Eradication of triple-negative breast cancer cells by targeting glycosylated PD-L1. Cancer Cell 33, 187–201.e10 (2018).

    Article  CAS  Google Scholar 

  23. Chen, G. et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 560, 382–386 (2018).

    Article  CAS  Google Scholar 

  24. Wang, H. et al. HIP1R targets PD-L1 to lysosomal degradation to alter T cell-mediated cytotoxicity. Nat. Chem. Biol. 15, 42–50 (2019).

    Article  CAS  Google Scholar 

  25. Chen, M. et al. Development and validation of a novel clinical fluorescence in situ hybridization assay to detect JAK2 and PD-L1 amplification: a fluorescence in situ hybridization assay for JAK2 and PD-L1 amplification. Mod. Pathol. 30, 1516–1526 (2017).

    Article  CAS  Google Scholar 

  26. Taguchi, T. & Misaki, R. Palmitoylation pilots Ras to recycling endosomes. Small GTPases 2, 82–84 (2011).

    Article  Google Scholar 

  27. Runkle, K. B. et al. Inhibition of DHHC20-mediated EGFR palmitoylation creates a dependence on EGFR signaling. Mol. Cell 62, 385–396 (2016).

    Article  CAS  Google Scholar 

  28. Gao, X. & Hannoush, R. N. Single-cell imaging of Wnt palmitoylation by the acyltransferase porcupine. Nat. Chem. Biol. 10, 61–68 (2014).

    Article  CAS  Google Scholar 

  29. Tukachinsky, H., Petrov, K., Watanabe, M. & Salic, A. Mechanism of inhibition of the tumor suppressor Patched by Sonic Hedgehog. Proc. Natl Acad. Sci. USA 113, E5866–E5875 (2016).

    Article  CAS  Google Scholar 

  30. Ren, J. et al. CSS-Palm 2.0: an updated software for palmitoylation sites prediction. Protein Eng. Des. Sel. 21, 639–644 (2008).

    Article  CAS  Google Scholar 

  31. Weng, S. L., Kao, H. J., Huang, C. H. & Lee, T. Y. MDD-Palm: identification of protein S-palmitoylation sites with substrate motifs based on maximal dependence decomposition. PLoS ONE 12, e0179529 (2017).

    Article  Google Scholar 

  32. Thul, P. J. & Lindskog, C. The human protein atlas: a spatial map of the human proteome. Protein Sci. 27, 233–244 (2018).

    Article  CAS  Google Scholar 

  33. Garcia-Diaz, A. et al. Interferon receptor signaling pathways regulating PD-L1 and PD-L2 expression. Cell Rep. 19, 1189–1201 (2017).

    Article  CAS  Google Scholar 

  34. Riaz, N. et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell 171, 934–949 e915 (2017).

    Article  CAS  Google Scholar 

  35. Linder, M. E. & Deschenes, R. J. Palmitoylation: policing protein stability and traffic. Nat. Rev. Mol. Cell Biol. 8, 74–84 (2007).

    Article  CAS  Google Scholar 

  36. Horita, H., Law, A., Hong, S. & Middleton, K. Identifying regulatory posttranslational modifications of PD-L1: a focus on monoubiquitinaton. Neoplasia 19, 346–353 (2017).

    Article  CAS  Google Scholar 

  37. Stringer, D. K. & Piper, R. C. A single ubiquitin is sufficient for cargo protein entry into MVBs in the absence of ESCRT ubiquitination. J. Cell Biol. 192, 229–242 (2011).

    Article  CAS  Google Scholar 

  38. Takahashi, H., Mayers, J. R., Wang, L., Edwardson, J. M. & Audhya, A. Hrs and STAM function synergistically to bind ubiquitin-modified cargoes in vitro. Biophys. J. 108, 76–84 (2015).

    Article  CAS  Google Scholar 

  39. Wang, H. et al. HIP1R targets PD-L1 to lysosomal degradation to alter T cell-mediated cytotoxicity. Nat. Chem. Biol. 15, 42–50 (2019).

    Article  CAS  Google Scholar 

  40. Bolhassani, A., Jafarzade, B. S. & Mardani, G. In vitro and in vivo delivery of therapeutic proteins using cell penetrating peptides. Peptides 87, 50–63 (2017).

    Article  CAS  Google Scholar 

  41. Soragni, A. et al. A designed inhibitor of p53 aggregation rescues p53 tumor suppression in ovarian carcinomas. Cancer Cell 29, 90–103 (2016).

    Article  CAS  Google Scholar 

  42. Liang, L. et al. A designed peptide targets two types of modifications of p53 with anti-cancer activity. Cell Chem. Biol. 25, 761–774.e5 (2018).

    Article  CAS  Google Scholar 

  43. Tian, X. et al. Organ-specific metastases obtained by culturing colorectal cancer cells on tissue-specific decellularized scaffolds. Nat. Biomed. Eng. 2, 443–452 (2018).

    Article  Google Scholar 

  44. Fang, C. et al. Identification of palmitoylated transitional endoplasmic reticulum ATPase by proteomic technique and pan antipalmitoylation antibody. J. Proteome Res. 15, 956–962 (2016).

    Article  CAS  Google Scholar 

  45. Yousefi-Salakdeh, E., Johansson, J. & Stromberg, R. A method for S- and O-palmitoylation of peptides: synthesis of pulmonary surfactant protein-C models. Biochem. J. 343, 557–562 (1999).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This project was supported by grants from the National Key Research and Development Plan (2016YFC0906000, 2016YFC0906003, 2016YFC0906002 and 2017YFC0906600), National Natural Science Foundation of China (81572326, 81322036, 81421001, 81773752, 81702969, 81874050, 21335002 and 31671360), Key Program of the Science and Technology Bureau of Sichuan (number 2017SZ00005), Top-Notch Young Talents Program of China (ZTZ2015-48), ‘Tang Scholar’ programme (JX-2017), Shanghai Municipal Education Commission-Gaofeng Clinical Medicine Grant Support (20152514), ‘ShuGuang’ project supported by the Shanghai Municipal Education Commission and Shanghai Education Development Foundation (15SG16), National Key Technology Support Program (2015BAI13B07 to J.X.), and Natural Science Foundation of Shanghai (18ZR1402800 to C.F.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

H.Y. performed the in vitro and in vivo experiments, analysed the data and wrote the manuscript. J.L. performed the in vivo experiments and analysed the data. C.L. performed the in vitro experiments. H.S. designed the in vivo experiments and analysed the data. J.-P.B. contributed to the experimental design, analysed the data and wrote the manuscript. H.W. performed the in vitro experiments. H.L. and C.F. contributed the anti-palmitoylation antibody and analysed the relevant data. Y.Z., L.L., X.Z. and C.W. analysed the peptide toxicity data in different organs/samples, as well as the tissue microarray data. Y.X. contributed analysis tools and designed the mutagenesis experiment. Y.C. analysed the peptide toxicity data. J.X. supervised the project, conceived and designed the experiments and peptides, analysed the data and wrote the manuscript.

Corresponding author

Correspondence to Jie Xu.

Ethics declarations

Competing interests

The 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.

Supplementary information

Supplementary Information

Supplementary methods, figures and tables

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yao, H., Lan, J., Li, C. et al. Inhibiting PD-L1 palmitoylation enhances T-cell immune responses against tumours. Nat Biomed Eng 3, 306–317 (2019). https://doi.org/10.1038/s41551-019-0375-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-019-0375-6

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