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:

A STING-activating nanovaccine for cancer immunotherapy

Abstract

The generation of tumour-specific T cells is critically important for cancer immunotherapy1,2. A major challenge in achieving a robust T-cell response is the spatiotemporal orchestration of antigen cross-presentation in antigen-presenting cells with innate stimulation. Here, we report a minimalist nanovaccine, comprising a simple physical mixture of an antigen and a synthetic polymeric nanoparticle, PC7A NP, which generates a strong cytotoxic T-cell response with low systemic cytokine expression. Mechanistically, the PC7A NP achieves efficient cytosolic delivery of tumour antigens to antigen-presenting cells in draining lymph nodes, leading to increased surface presentation while simultaneously activating type I interferon-stimulated genes. This effect is dependent on stimulator of interferon genes (STING), but not the Toll-like receptor or the mitochondrial antiviral-signalling protein (MAVS) pathway. The nanovaccine led to potent tumour growth inhibition in melanoma, colon cancer and human papilloma virus-E6/E7 tumour models. The combination of the PC7A nanovaccine and an anti-PD-1 antibody showed great synergy, with 100% survival over 60 days in a TC-1 tumour model. Rechallenging of these tumour-free animals with TC-1 cells led to complete inhibition of tumour growth, suggesting the generation of long-term antitumour memory. The STING-activating nanovaccine offers a simple, safe and robust strategy in boosting anti-tumour immunity for cancer immunotherapy.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: PC7A NPs induce robust antigen-specific CTL, Th1 and Th2 responses.
Figure 2: PC7A NPs improve antigen delivery and cross-presentation in APCs and stimulate CD8 T-cell responses.
Figure 3: PC7A NPs activate APCs in draining lymph nodes and stimulate STING-dependent adaptive immune responses.
Figure 4: PC7A nanovaccine inhibits tumour growth and prolongs survival in tumour-bearing mice.

Similar content being viewed by others

References

  1. Rosenberg, S. A. & Restifo, N. P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348, 62–68 (2015).

    Article  CAS  Google Scholar 

  2. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).

    Article  CAS  Google Scholar 

  3. Kuai, R., Ochyl, L. J., Bahjat, K. S., Schwendeman, A. & Moon, J. J. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nat. Mater. 16, 489–496 (2017).

    Article  CAS  Google Scholar 

  4. Liu, H. et al. Structure-based programming of lymph-node targeting in molecular vaccines. Nature 507, 519–522 (2014).

    Article  CAS  Google Scholar 

  5. Reddy, S. T. et al. Exploiting lymphatic transport and complement activation in nanoparticle vaccines. Nat. Biotechnol. 25, 1159–1164 (2007).

    Article  CAS  Google Scholar 

  6. Ma, X. et al. Ultra-pH-sensitive nanoprobe library with broad pH tunability and fluorescence emissions. J. Am. Chem. Soc. 136, 11085–11092 (2014).

    Article  CAS  Google Scholar 

  7. Wang, C. et al. A nanobuffer reporter library for fine-scale imaging and perturbation of endocytic organelles. Nat. Commun. 6, 8524 (2015).

    Article  CAS  Google Scholar 

  8. Boussif, O. et al. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc. Natl Acad. Sci. USA 92, 7297–7301 (1995).

    Article  CAS  Google Scholar 

  9. Maldonado, R. A. et al. Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc. Natl Acad. Sci. USA 112, E156–E165 (2015).

    Article  CAS  Google Scholar 

  10. Poltorak, A. et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science 282, 2085–2088 (1998).

    Article  CAS  Google Scholar 

  11. Hemmi, H. et al. A toll-like receptor recognizes bacterial DNA. Nature 408, 740–745 (2000).

    Article  CAS  Google Scholar 

  12. Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8α+ dendritic cells in cytotoxic T cell immunity. Science 322, 1097–1100 (2008).

    Article  CAS  Google Scholar 

  13. Heath, W. R. et al. Cross-presentation, dendritic cell subsets, and the generation of immunity to cellular antigens. Immunol. Rev. 199, 9–26 (2004).

    Article  CAS  Google Scholar 

  14. Wilson, J. T. et al. pH-responsive nanoparticle vaccines for dual-delivery of antigens and immunostimulatory oligonucleotides. ACS Nano 7, 3912–3925 (2013).

    Article  CAS  Google Scholar 

  15. Wang, Z. et al. A redox-activatable fluorescent sensor for the high-throughput quantification of cytosolic delivery of macromolecules. Angew. Chem. Int. Ed. 56, 1319–1323 (2017).

    Article  CAS  Google Scholar 

  16. Liechtenstein, T., Dufait, I., Lanna, A., Breckpot, K. & Escors, D. Modulating co-stimulation during antigen presentation to enhance cancer immunotherapy. Immunol. Endocr. Metab. Agents Med. Chem. 12, 224–235 (2012).

    Article  CAS  Google Scholar 

  17. Zitvogel, L., Galluzzi, L., Kepp, O., Smyth, M. J. & Kroemer, G. Type I interferons in anticancer immunity. Nat. Rev. Immunol. 15, 405–414 (2015).

    Article  CAS  Google Scholar 

  18. Fuertes, M. B., Woo, S. R., Burnett, B., Fu, Y. X. & Gajewski, T. F. Type I interferon response and innate immune sensing of cancer. Trends Immunol. 34, 67–73 (2013).

    Article  CAS  Google Scholar 

  19. Trinchieri, G. Type I interferon: friend or foe? J. Exp. Med. 207, 2053–2063 (2010).

    Article  CAS  Google Scholar 

  20. Alexopoulou, L., Holt, A. C., Medzhitov, R. & Flavell, R. A. Recognition of double-stranded RNA and activation of NF-κB by Toll-like receptor 3. Nature 413, 732–738 (2001).

    Article  CAS  Google Scholar 

  21. Baccala, R., Hoebe, K., Kono, D. H., Beutler, B. & Theofilopoulos, A. N. TLR-dependent and TLR-independent pathways of type I interferon induction in systemic autoimmunity. Nat. Med. 13, 543–551 (2007).

    Article  CAS  Google Scholar 

  22. Sun, L., Wu, J., Du, F., Chen, X. & Chen, Z. J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339, 786–791 (2013).

    Article  CAS  Google Scholar 

  23. Carroll, E. C. et al. The vaccine adjuvant chitosan promotes cellular immunity via DNA sensor cGAS-STING-dependent induction of type I interferons. Immunity 44, 597–608 (2016).

    Article  CAS  Google Scholar 

  24. Woo, S. R. et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity 41, 830–842 (2014).

    Article  CAS  Google Scholar 

  25. Lemos, H., Huang, L., McGaha, T. L. & Mellor, A. L. Cytosolic DNA sensing via the stimulator of interferon genes adaptor: Yin and Yang of immune responses to DNA. Eur. J. Immunol. 44, 2847–2853 (2014).

    Article  CAS  Google Scholar 

  26. Kreiter, S. et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 520, 692–696 (2015).

    Article  CAS  Google Scholar 

  27. Yadav, M. et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature 515, 572–576 (2014).

    CAS  Google Scholar 

  28. Sun, Y. Y. et al. Local HPV recombinant vaccinia boost following priming with an HPV DNA vaccine enhances local HPV-specific CD8+ T cell mediated tumor control in the genital tract. Clin. Cancer Res. 22, 657–669 (2016).

    Article  CAS  Google Scholar 

  29. Liu, Z., Zhou, H., Wang, W., Fu, Y. X. & Zhu, M. A novel dendritic cell targeting HPV16 E7 synthetic vaccine in combination with PD-L1 blockade elicits therapeutic antitumor immunity in mice. Oncoimmunology 5, e1147641 (2016).

    Article  Google Scholar 

  30. Rice, A. E. et al. An HPV-E6/E7 immunotherapy plus PD-1 checkpoint inhibition results in tumor regression and reduction in PD-L1 expression. Cancer Gene. Ther. 22, 454–462 (2015).

    Article  CAS  Google Scholar 

  31. Holmgaard, R. B., Zamarin, D., Munn, D. H., Wolchok, J. D. & Allison, J. P. Indoleamine 2,3-dioxygenase is a critical resistance mechanism in antitumor T cell immunotherapy targeting CTLA-4. J. Exp. Med. 210, 1389–1402 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  33. Sharma, P. & Allison, J. P. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 161, 205–214 (2015).

    Article  CAS  Google Scholar 

  34. Zhou, K. et al. Tunable, ultrasensitive pH-responsive nanoparticles targeting specific endocytic organelles in living cells. Angew. Chem. Int. Ed. 50, 6109–6114 (2011).

    Article  CAS  Google Scholar 

  35. Zhou, K. et al. Multicolored pH-tunable and activatable fluorescence nanoplatform responsive to physiologic pH stimuli. J. Am. Chem. Soc. 134, 7803–7811 (2012).

    Article  CAS  Google Scholar 

  36. Tsarevsky, N. V. & Matyjaszewski, K. ‘Green’ atom transfer radical polymerization: from process design to preparation of well-defined environmentally friendly polymeric materials. Chem. Rev. 107, 2270–2299 (2007).

    Article  CAS  Google Scholar 

  37. Li, X. D. et al. Pivotal roles of cGAS-cGAMP signaling in antiviral defense and immune adjuvant effects. Science 341, 1390–1394 (2013).

    Article  CAS  Google Scholar 

  38. Collins, A. C. et al. Cyclic GMP-AMP synthase is an innate immune DNA sensor for Mycobacterium tuberculosis. Cell Host Microbe. 17, 820–828 (2015).

    Article  CAS  Google Scholar 

  39. Zhang, X. et al. Cyclic GMP-AMP containing mixed phosphodiester linkages is an endogenous high-affinity ligand for STING. Mol. Cell 51, 226–235 (2013).

    Article  CAS  Google Scholar 

  40. Huang, L. et al. Engineering DNA nanoparticles as immunomodulatory reagents that activate regulatory T cells. J. Immunol. 188, 4913–4920 (2012).

    Article  CAS  Google Scholar 

  41. Hoshi, M. et al. The absence of IDO upregulates type I IFN production, resulting in suppression of viral replication in the retrovirus-infected mouse. J. Immunol. 185, 3305–3312 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the National Institutes of Health (R01AI093967 to Z.J.C., R01EB013149 and R01CA192221 to J.G.) and the Cancer Prevention and Research Institute of Texas (RP120718-P3 and RP150498 to Z.J.C.). Z.J.C. is an Investigator of the Howard Hughes Medical Institute. M.R.P. is a Dedman Scholar in Clinical Care. Animal imaging work is supported by a UT Southwestern Small Animal Imaging Resource Grant (U24 CA126608) and a Simmons Cancer Center Support Grant (P30 CA142543). The authors thank Z. Zeng for cell culture, Q. Wei for polymer synthesis and T. Zhao for animal imaging. The authors also thank N. Yan for discussions on STING activation, T.C. Wu for providing the TC-1 tumour cells, P. Hwu for the B16-OVA cancer cells, Y. Peng for vaccine safety analysis, S. Tso for ITC experiments and the molecular pathology core of UT Southwestern for tissue toxicity analysis.

Author information

Authors and Affiliations

Authors

Contributions

M.L. and Z.W. designed and performed the majority of experiments, analysed the data and wrote the first draft of the paper. H.W. made the initial observation of the immune stimulatory activity of PC7A and performed the experiments on CD8 T cell activation by PC7A in vitro and in vivo. H.C. analysed innate cytokine expression in local tissues and cell lines. Z.L. assisted with the CTL and Th1 experiments. Y.L. and M.D. assisted with the PC7A and STING interaction experiments. G.H. and C.W. assisted with animal efficacy evaluation. X.C. assisted with transgenic mice studies. Y.F. assisted with PD-L1 expression in B16 and TC-1 tumours. M.R.P., J.L. and A.E.F. contributed to experimental designs. Z.J.C. and J.G. supervised all the experiments and revised the final manuscript.

Corresponding authors

Correspondence to Zhijian J. Chen or Jinming Gao.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary information

Supplementary information (PDF 3198 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Luo, M., Wang, H., Wang, Z. et al. A STING-activating nanovaccine for cancer immunotherapy. Nature Nanotech 12, 648–654 (2017). https://doi.org/10.1038/nnano.2017.52

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nnano.2017.52

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing