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:

Close the cancer–immunity cycle by integrating lipid nanoparticle–mRNA formulations and dendritic cell therapy

Abstract

Effective cancer immunotherapy is usually blocked by immunosuppressive factors in the tumour microenvironment, resulting in tumour promotion, metastasis and recurrence. Here we combine lipid nanoparticle–mRNA formulations and dendritic cell therapy (named CATCH) to boost the cancer–immunity cycle via progressive steps to overcome the immunosuppressive tumour microenvironment. Multiple types of sugar-alcohol-derived lipid nanoparticles are conceived to modulate the cancer–immunity cycle. First, one type of lipid nanoparticle containing CD40 ligand mRNA induces robust immunogenic cell death in tumoural tissues, leading to the release of tumour-associated antigens and the expression of CD40 ligand. Next, dendritic cells engineered by another type of lipid nanoparticle encapsulating CD40 mRNA are adoptively transferred, which are then activated by the CD40 ligand molecules in tumoural tissues. This promotes the secretion of multiple cytokines and chemokines, and the upregulation of co-stimulatory molecules on dendritic cells, which are crucial for reprogramming the tumour microenvironment and priming the T-cell responses. After dendritic cells present tumour-associated antigens to T cells, all the above stepwise events contribute to boosting a potent tumour-specific T-cell immunity that eradicates established tumours, suppresses distal lesions and prevents tumour rechallenge.

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: Chemical synthesis of sugar-alcohol-derived ionizable lipids.
Fig. 2: Screening, optimization and characterization of LNP–mRNA formulations.
Fig. 3: DC activation, LNP-induced ICD and therapeutic effects on primary tumour and rechallenged tumour.
Fig. 4: Therapeutic effects on two tumour or immune cell depletion models, and dynamic expression of cytokines and chemokines in tumoural tissues and blood.
Fig. 5: Changes in immune cell infiltration, memory T cells and mRNA transcripts.
Fig. 6: Applicability of CATCH treatment regimen in other tumour models and human-derived cells.

Similar content being viewed by others

Data availability

Source data are provided with this paper. Additional materials for this study are available from the corresponding author upon reasonable request.

References

  1. Chen, D. S. & Mellman, I. Oncology meets immunology: the cancer-immunity cycle. Immunity 39, 1–10 (2013).

    Google Scholar 

  2. Wculek, S. K. et al. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 20, 7–24 (2020).

    CAS  Google Scholar 

  3. Anguille, S., Smits, E. L., Lion, E., van Tendeloo, V. F. & Berneman, Z. N. Clinical use of dendritic cells for cancer therapy. Lancet Oncol. 15, e257–e267 (2014).

    CAS  Google Scholar 

  4. Belderbos, R. A., Aerts, J. G. & Vroman, H. Enhancing dendritic cell therapy in solid tumors with immunomodulating conventional treatment. Mol. Ther. Oncolytics 13, 67–81 (2019).

    CAS  Google Scholar 

  5. Saxena, M., van der Burg, S. H., Melief, C. J. & Bhardwaj, N. Therapeutic cancer vaccines. Nat. Rev. Cancer 21, 360–378 (2021).

    CAS  Google Scholar 

  6. Spranger, S., Bao, R. & Gajewski, T. F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 523, 231–235 (2015).

    CAS  Google Scholar 

  7. Tang, M. et al. Toll-like receptor 2 activation promotes tumor dendritic cell dysfunction by regulating IL-6 and IL-10 receptor signaling. Cell Rep. 13, 2851–2864 (2015).

    CAS  Google Scholar 

  8. Villablanca, E. J. et al. Tumor-mediated liver X receptor-α activation inhibits CC chemokine receptor-7 expression on dendritic cells and dampens antitumor responses. Nat. Med. 16, 98–105 (2010).

    CAS  Google Scholar 

  9. Veglia, F. et al. Lipid bodies containing oxidatively truncated lipids block antigen cross-presentation by dendritic cells in cancer. Nat. Commun. 8, 2122 (2017).

    Google Scholar 

  10. Gottfried, E. et al. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood 107, 2013–2021 (2006).

    CAS  Google Scholar 

  11. Park, M. D. et al. On the biology and therapeutic modulation of macrophages and dendritic cells in cancer. Annu. Rev. Cancer Biol. 7, 291–311 (2023).

    Google Scholar 

  12. Merad, M., Sathe, P., Helft, J., Miller, J. & Mortha, A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu. Rev. Immunol. 31, 563–604 (2013).

    CAS  Google Scholar 

  13. Harari, A., Graciotti, M., Bassani-Sternberg, M. & Kandalaft, L. E. Antitumour dendritic cell vaccination in a priming and boosting approach. Nat. Rev. Drug Discov. 19, 635–652 (2020).

    CAS  Google Scholar 

  14. Kongsted, P. et al. Dendritic cell vaccination in combination with docetaxel for patients with metastatic castration-resistant prostate cancer: a randomized phase II study. Cytotherapy 19, 500–513 (2017).

    CAS  Google Scholar 

  15. Tanyi, J. L. et al. Personalized cancer vaccine effectively mobilizes antitumor T cell immunity in ovarian cancer. Sci. Transl. Med. 10, eaao5931 (2018).

    Google Scholar 

  16. Saxena, M., Balan, S., Roudko, V. & Bhardwaj, N. Towards superior dendritic-cell vaccines for cancer therapy. Nat. Biomed. Eng. 2, 341–346 (2018).

    Google Scholar 

  17. Kroemer, G., Galluzzi, L., Kepp, O. & Zitvogel, L. Immunogenic cell death in cancer therapy. Annu. Rev. Immunol. 31, 51–72 (2013).

    CAS  Google Scholar 

  18. Galluzzi, L., Buqué, A., Kepp, O., Zitvogel, L. & Kroemer, G. Immunogenic cell death in cancer and infectious disease. Nat. Rev. Immunol. 17, 97–111 (2017).

    CAS  Google Scholar 

  19. Bhardwaj, N. et al. Flt3 ligand augments immune responses to anti-DEC-205-NY-ESO-1 vaccine through expansion of dendritic cell subsets. Nat. Cancer 1, 1204–1217 (2020).

    CAS  Google Scholar 

  20. De Keersmaecker, B. et al. TriMix and tumor antigen mRNA electroporated dendritic cell vaccination plus ipilimumab: link between T-cell activation and clinical responses in advanced melanoma. J. Immunother. Cancer 8, e000329 (2020).

    Google Scholar 

  21. Lau, S. P. et al. Dendritic cell vaccination and CD40-agonist combination therapy licenses T cell-dependent antitumor immunity in a pancreatic carcinoma murine model. J. Immunother. Cancer 8, e000772 (2020).

    Google Scholar 

  22. Vonderheide, R. H. CD40 agonist antibodies in cancer immunotherapy. Annu. Rev. Med. 71, 47–58 (2020).

    CAS  Google Scholar 

  23. Bullock, T. N. CD40 stimulation as a molecular adjuvant for cancer vaccines and other immunotherapies. Cell. Mol. Immunol. 19, 14–22 (2022).

    CAS  Google Scholar 

  24. Salomon, R. et al. Bispecific antibodies increase the therapeutic window of CD40 agonists through selective dendritic cell targeting. Nat. Cancer 3, 287–302 (2022).

    CAS  Google Scholar 

  25. Li, D.-K. & Wang, W. Characteristics and clinical trial results of agonistic anti‑CD40 antibodies in the treatment of malignancies. Oncol. Lett. 20, 176 (2020).

    CAS  Google Scholar 

  26. Beatty, G. L. et al. A phase I study of an agonist CD40 monoclonal antibody (CP-870,893) in combination with gemcitabine in patients with advanced pancreatic ductal adenocarcinoma. Clin. Cancer Res. 19, 6286–6295 (2013).

    CAS  Google Scholar 

  27. Vonderheide, R. H. et al. Phase I study of the CD40 agonist antibody CP-870,893 combined with carboplatin and paclitaxel in patients with advanced solid tumors. Oncoimmunology 2, e23033 (2013).

    Google Scholar 

  28. Nowak, A. K. et al. A phase 1b clinical trial of the CD40-activating antibody CP-870,893 in combination with cisplatin and pemetrexed in malignant pleural mesothelioma. Ann. Oncol. 26, 2483–2490 (2015).

    CAS  Google Scholar 

  29. Aricò, F., Tundo, P., Maranzana, A. & Tonachini, G. Synthesis of five‐membered cyclic ethers by reaction of 1,4‐diols with dimethyl carbonate. ChemSusChem 5, 1578–1586 (2012).

    Google Scholar 

  30. Sahay, G., Alakhova, D. Y. & Kabanov, A. V. Endocytosis of nanomedicines. J. Control. Release 145, 182–195 (2010).

    CAS  Google Scholar 

  31. Hou, X. et al. Vitamin lipid nanoparticles enable adoptive macrophage transfer for the treatment of multidrug-resistant bacterial sepsis. Nat. Nanotechnol. 15, 41–46 (2020).

    CAS  Google Scholar 

  32. Li, Y. et al. Multifunctional oncolytic nanoparticles deliver self-replicating IL-12 RNA to eliminate established tumors and prime systemic immunity. Nat. Cancer 1, 882–893 (2020).

    CAS  Google Scholar 

  33. Kikuchi, T., Moore, M. A. & Crystal, R. G. Dendritic cells modified to express CD40 ligand elicit therapeutic immunity against preexisting murine tumors. Blood 96, 91–99 (2000).

    CAS  Google Scholar 

  34. Hernandez, M. G. H., Shen, L. & Rock, K. L. CD40-CD40 ligand interaction between dendritic cells and CD8+ T cells is needed to stimulate maximal T cell responses in the absence of CD4+ T cell help. J. Immunol. 178, 2844–2852 (2007).

    CAS  Google Scholar 

  35. Love, S. A., Maurer-Jones, M. A., Thompson, J. W., Lin, Y.-S. & Haynes, C. L. Assessing nanoparticle toxicity. Annu. Rev. Anal. Chem. 5, 181–205 (2012).

    CAS  Google Scholar 

  36. Yang, W., Wang, L., Mettenbrink, E. M., DeAngelis, P. L. & Wilhelm, S. Nanoparticle toxicology. Annu. Rev. Pharmacol. Toxicol. 61, 269–289 (2021).

    CAS  Google Scholar 

  37. Melero, I., Castanon, E., Alvarez, M., Champiat, S. & Marabelle, A. Intratumoural administration and tumour tissue targeting of cancer immunotherapies. Nat. Rev. Clin. Oncol. 18, 558–576 (2021).

    CAS  Google Scholar 

  38. Hewitt, S. L. et al. Durable anticancer immunity from intratumoral administration of IL-23, IL-36γ, and OX40L mRNAs. Sci. Transl. Med. 11, eaat9143 (2019).

    CAS  Google Scholar 

  39. Hotz, C. et al. Local delivery of mRNA-encoded cytokines promotes antitumor immunity and tumor eradication across multiple preclinical tumor models. Sci. Transl. Med. 13, eabc7804 (2021).

    CAS  Google Scholar 

  40. Murray, P. J. et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41, 14–20 (2014).

    CAS  Google Scholar 

  41. Jhunjhunwala, S., Hammer, C. & Delamarre, L. Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat. Rev. Cancer 21, 298–312 (2021).

    CAS  Google Scholar 

  42. Chen, L. & Flies, D. B. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat. Rev. Immunol. 13, 227–242 (2013).

    Google Scholar 

  43. Mueller, S. N., Gebhardt, T., Carbone, F. R. & Heath, W. R. Memory T cell subsets, migration patterns, and tissue residence. Annu. Rev. Immunol. 31, 137–161 (2013).

    CAS  Google Scholar 

  44. Sagiv-Barfi, I. et al. Therapeutic antitumor immunity by checkpoint blockade is enhanced by ibrutinib, an inhibitor of both BTK and ITK. Proc. Natl Acad. Sci. USA 112, E966–E972 (2015).

    CAS  Google Scholar 

  45. Ouzounova, M. et al. Monocytic and granulocytic myeloid derived suppressor cells differentially regulate spatiotemporal tumour plasticity during metastatic cascade. Nat. Commun. 8, 14979 (2017).

    CAS  Google Scholar 

  46. Zeng, C. et al. Leveraging mRNA sequences and nanoparticles to deliver SARS‐CoV‐2 antigens in vivo. Adv. Mater. 32, 2004452 (2020).

    CAS  Google Scholar 

  47. Li, W. et al. Biomimetic nanoparticles deliver mRNAs encoding costimulatory receptors and enhance T cell mediated cancer immunotherapy. Nat. Commun. 12, 7264 (2021).

    CAS  Google Scholar 

  48. Li, B. et al. An orthogonal array optimization of lipid-like nanoparticles for mRNA delivery in vivo. Nano Lett. 15, 8099–8107 (2015).

    CAS  Google Scholar 

Download references

Acknowledgements

Cryo-TEM imaging was performed at the Center for Electron Microscopy and Analysis (CEMAS) at The Ohio State University. The Genomics Shared Resource performed the nCounter NanoString analysis for this study with part support from the National Cancer Institute (grant P30 CA016058). Y.D. acknowledges support from the Maximizing Investigators’ Research Award R35GM144117 from the National Institute of General Medical Sciences as well as the funding from the Icahn School of Medicine at Mount Sinai. X.H. and J.Y. acknowledge support from the Professor Sylvan G. Frank Graduate Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

Y. Zhang, X.H. and S.D. conceived the work, performed the experiments, analysed the data and wrote the paper. Y.X., J.Y., D.D.K., Y. Zhong and C.W. contributed to the mRNA synthesis, LNP characterization and flow cytometry assays. B.D. and D.W.M. contributed to the cryo-TEM imaging. Y.D. conceived and supervised the project and wrote the paper. The final paper was edited and approved by all authors.

Corresponding author

Correspondence to Yizhou Dong.

Ethics declarations

Competing interests

Y.D. is a scientific advisory board member of Oncorus Inc., Arbor Biotechnologies and FL85. The other authors declare no competing interests.

Peer review

Peer review information

Nature Nanotechnology thanks the anonymous reviewers 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.

Supplementary information

Supplementary Information

Supplementary Figs. 1–10, Methods, Discussion and References.

Reporting Summary

Supplementary Data 1

Source data for supplementary figures.

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. 5

Statistical source data.

Source Data Fig. 6

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

Zhang, Y., Hou, X., Du, S. et al. Close the cancer–immunity cycle by integrating lipid nanoparticle–mRNA formulations and dendritic cell therapy. Nat. Nanotechnol. 18, 1364–1374 (2023). https://doi.org/10.1038/s41565-023-01453-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41565-023-01453-9

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research