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:

Targeting vaccines to dendritic cells by mimicking the processing and presentation of antigens in xenotransplant rejection

Abstract

Targeting the delivery of vaccines to dendritic cells (DCs) is challenging. Here we show that, by mimicking the fast and strong antigen processing and presentation that occurs during the rejection of xenotransplanted tissue, xenogeneic cell membrane-derived vesicles exposing tissue-specific antibodies can be leveraged to deliver peptide antigens and mRNA-encoded antigens to DCs. In mice with murine melanoma and murine thymoma, xenogeneic vesicles encapsulating a tumour-derived antigenic peptide or coated on lipid nanoparticles encapsulating an mRNA coding for a tumour antigen elicited potent tumour-specific T-cell responses that inhibited tumour growth. Mice immunized with xenogeneic vesicle-coated lipid nanoparticles encapsulating an mRNA encoding for the spike protein of severe acute respiratory syndrome coronavirus 2 elicited titres of anti-spike receptor-binding domain immunoglobulin G and of neutralizing antibodies that were approximately 32-fold and 6-fold, respectively, those elicited by a commercialized mRNA–lipid nanoparticle vaccine. The advantages of mimicking the biological recognition between immunoglobulin G on xenogeneic vesicles and fragment crystallizable receptors on DCs may justify the assessment of the safety risks of using animal-derived biological products in humans.

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: IgG binding donates XMV DC targeting capacity.
Fig. 2: XMVs enhance DC maturation and cross-presentation.
Fig. 3: XMVs deliver antigen peptide specifically to LNs and elicit potent immune responses.
Fig. 4: XMV–Ag induces potent antitumour T-cell responses.
Fig. 5: XMV coating on LNP delivering an mRNA-encoding antigen for cancer and COVID-19 vaccination.

Similar content being viewed by others

Data availability

The data supporting the results in this study are available within the paper and its Supplementary Information. The raw and analysed datasets generated during the study are available for research purposes from the corresponding authors on reasonable request. Source data are provided with this paper.

References

  1. Rappuoli, R., Mandl, C. W., Black, S. & De Gregorio, E. Vaccines for the twenty-first century society. Nat. Rev. Immunol. 11, 865–872 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Chen, S. et al. Nanotechnology-based mRNA vaccines. Nat. Rev. Methods Primers 3, 63 (2023).

    CAS  Google Scholar 

  3. Barbier, A. J., Jiang, A. Y., Zhang, P., Wooster, R. & Anderson, D. G. The clinical progress of mRNA vaccines and immunotherapies. Nat. Biotechnol. 40, 840–854 (2022).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  5. Steinman, R. M. & Banchereau, J. Taking dendritic cells into medicine. Nature 449, 419–426 (2007).

    CAS  PubMed  Google Scholar 

  6. Pulendran, B. & Ahmed, R. Immunological mechanisms of vaccination. Nat. Immunol. 12, 509–517 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Pollard, A. J. & Bijker, E. M. A guide to vaccinology: from basic principles to new developments. Nat. Rev. Immunol. 21, 83–100 (2021).

    CAS  PubMed  Google Scholar 

  8. De Koker, S. et al. Designing polymeric particles for antigen delivery. Chem. Soc. Rev. 40, 320–339 (2011).

    PubMed  Google Scholar 

  9. Guerrini, G., Magrì, D., Gioria, S., Medaglini, D. & Calzolai, L. Characterization of nanoparticles-based vaccines for COVID-19. Nat. Nanotechnol. 17, 570–576 (2022).

    CAS  PubMed  Google Scholar 

  10. Baden, L. R. et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N. Engl. J. Med. 384, 403–416 (2021).

    CAS  PubMed  Google Scholar 

  11. Chaudhary, N., Weissman, D. & Whitehead, K. A. mRNA vaccines for infectious diseases: principles, delivery and clinical translation. Nat. Rev. Drug Discov. 20, 817–838 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Wang, Z. et al. Exosomes decorated with a recombinant SARS-CoV-2 receptor-binding domain as an inhalable COVID-19 vaccine. Nat. Biomed. Eng. 6, 791–805 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Kim, J. et al. Injectable, spontaneously assembling, inorganic scaffolds modulate immune cells in vivo and increase vaccine efficacy. Nat. Biotechnol. 33, 64–72 (2015).

    CAS  PubMed  Google Scholar 

  14. Huang, W.-C. et al. A malaria vaccine adjuvant based on recombinant antigen binding to liposomes. Nat. Nanotechnol. 13, 1174–1181 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. He, L. et al. Presenting native-like trimeric HIV-1 antigens with self-assembling nanoparticles. Nat. Commun. 7, 12041 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Gong, N. et al. Proton-driven transformable nanovaccine for cancer immunotherapy. Nat. Nanotechnol. 15, 1053–1064 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Yin, Q. et al. A TLR7-nanoparticle adjuvant promotes a broad immune response against heterologous strains of influenza and SARS-CoV-2. Nat. Mater. 22, 380–390 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Lee, J. et al. In vivo fate and intracellular trafficking of vaccine delivery systems. Adv. Drug Deliv. Rev. 186, 114325 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Rohner, E., Yang, R., Foo, K. S., Goedel, A. & Chien, K. R. Unlocking the promise of mRNA therapeutics. Nat. Biotechnol. 40, 1586–1600 (2022).

    CAS  PubMed  Google Scholar 

  20. Chen, J. et al. Lipid nanoparticle-mediated lymph node-targeting delivery of mRNA cancer vaccine elicits robust CD8+ T cell response. Proc. Natl Acad. Sci. USA 119, e2207841119 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Krauson, A. J., Casimero, F. V. C., Siddiquee, Z. & Stone, J. R. Duration of SARS-CoV-2 mRNA vaccine persistence and factors associated with cardiac involvement in recently vaccinated patients. npj Vaccines 8, 141 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Lee, J., Woodruff, M. C., Kim, E. H. & Nam, J.-H. Knife’s edge: balancing immunogenicity and reactogenicity in mRNA vaccines. Exp. Mol. Med. 55, 1305–1313 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Trougakos, I. P. et al. COVID-19 mRNA vaccine-induced adverse effects: unwinding the unknowns. Trends Mol. Med. 28, 800–802 (2022).

    PubMed  PubMed Central  Google Scholar 

  24. Zin Tun, G. S., Gleeson, D., Al-Joudeh, A. & Dube, A. Immune-mediated hepatitis with the Moderna vaccine, no longer a coincidence but confirmed. J. Hepatol. 76, 747–749 (2022).

    CAS  PubMed  Google Scholar 

  25. Bachmann, M. F. & Jennings, G. T. Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns. Nat. Rev. Immunol. 10, 787–796 (2010).

    CAS  PubMed  Google Scholar 

  26. Wang, W. et al. Dual-targeting nanoparticle vaccine elicits a therapeutic antibody response against chronic hepatitis B. Nat. Nanotechnol. 15, 406–416 (2020).

    PubMed  PubMed Central  Google Scholar 

  27. Xia, Y. et al. Exploiting the pliability and lateral mobility of Pickering emulsion for enhanced vaccination. Nat. Mater. 17, 187–194 (2018).

    CAS  PubMed  Google Scholar 

  28. 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).

    CAS  PubMed  Google Scholar 

  29. Liu, C. et al. A nanovaccine for antigen self-presentation and immunosuppression reversal as a personalized cancer immunotherapy strategy. Nat. Nanotechnol. 17, 531–540 (2022).

    CAS  PubMed  Google Scholar 

  30. Conniot, J. et al. Immunization with mannosylated nanovaccines and inhibition of the immune-suppressing microenvironment sensitizes melanoma to immune checkpoint modulators. Nat. Nanotechnol. 14, 891–901 (2019).

    CAS  PubMed  Google Scholar 

  31. 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  PubMed  Google Scholar 

  32. Wang, H. et al. Metabolic labeling and targeted modulation of dendritic cells. Nat. Mater. 19, 1244–1252 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Bros, M. et al. The protein corona as a confounding variable of nanoparticle-mediated targeted vaccine delivery. Front. Immunol. 9, 1760 (2018).

    PubMed  PubMed Central  Google Scholar 

  34. Mo, J., Xie, Q., Wei, W. & Zhao, J. Revealing the immune perturbation of black phosphorus nanomaterials to macrophages by understanding the protein corona. Nat. Commun. 9, 2480 (2018).

    PubMed  PubMed Central  Google Scholar 

  35. Vu, V. P. et al. Immunoglobulin deposition on biomolecule corona determines complement opsonization efficiency of preclinical and clinical nanoparticles. Nat. Nanotechnol. 14, 260–268 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  37. Du, Y., Zhou, H., Su, G., Ma, M. & Liu, Y. Protein corona-driven nanovaccines improve antigen intracellular release and immunotherapy efficacy. J. Control. Release 345, 601–609 (2022).

    CAS  PubMed  Google Scholar 

  38. Sykes, M. & Sachs, D. H. Progress in xenotransplantation: overcoming immune barriers. Nat. Rev. Nephrol. 18, 745–761 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Jaycox, J. R., Dai, Y. & Ring, A. M. Decoding the autoantibody reactome. Science 383, 705–707 (2024).

    CAS  PubMed  Google Scholar 

  40. Anand, R. P. et al. Design and testing of a humanized porcine donor for xenotransplantation. Nature 622, 393–401 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Baker, K., Rath, T., Lencer, W. I., Fiebiger, E. & Blumberg, R. S. Cross-presentation of IgG-containing immune complexes. Cell. Mol. Life Sci. 70, 1319–1334 (2013).

    CAS  PubMed  Google Scholar 

  42. Baker, K. et al. Neonatal Fc receptor for IgG (FcRn) regulates cross-presentation of IgG immune complexes by CD8CD11b+ dendritic cells. Proc. Natl Acad. Sci. USA 108, 9927–9932 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Giodini, A., Rahner, C. & Cresswell, P. Receptor-mediated phagocytosis elicits cross-presentation in nonprofessional antigen-presenting cells. Proc. Natl Acad. Sci. USA 106, 3324–3329 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Clatworthy, M. R. et al. Immune complexes stimulate CCR7-dependent dendritic cell migration to lymph nodes. Nat. Med. 20, 1458–1463 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Rogers, N. J., Mirenda, V., Jackson, I., Dorling, A. & Lechler, R. I. Costimulatory blockade by the induction of an endogenous xenospecific antibody response. Nat. Immunol. 1, 163–168 (2000).

    CAS  PubMed  Google Scholar 

  46. Wu, F. et al. Fluorescence imaging of the lymph node uptake of proteins in mice after subcutaneous injection: molecular weight dependence. Pharm. Res. 29, 1843–1853 (2012).

    CAS  PubMed  Google Scholar 

  47. Roseboom, I. C., Rosing, H., Beijnen, J. H. & Dorlo, T. P. C. Skin tissue sample collection, sample homogenization, and analyte extraction strategies for liquid chromatographic mass spectrometry quantification of pharmaceutical compounds. J. Pharm. Biomed. Anal. 191, 113590 (2020).

    CAS  PubMed  Google Scholar 

  48. Hu, C.-M. J. et al. Nanoparticle biointerfacing by platelet membrane cloaking. Nature 526, 118–121 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Fang, R. H., Kroll, A. V., Gao, W. & Zhang, L. Cell membrane coating nanotechnology. Adv. Mater. 30, 1706759 (2018).

    Google Scholar 

  50. Zhai, Y. et al. T lymphocyte membrane-decorated epigenetic nanoinducer of interferons for cancer immunotherapy. Nat. Nanotechnol. 16, 1271–1280 (2021).

    CAS  PubMed  Google Scholar 

  51. Elechalawar, C. K., Hossen, M. N., McNally, L., Bhattacharya, R. & Mukherjee, P. Analysing the nanoparticle–protein corona for potential molecular target identification. J. Control. Release 322, 122–136 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Brasel, K., De Smedt, T., Smith, J. L. & Maliszewski, C. R. Generation of murine dendritic cells from flt3-ligand-supplemented bone marrow cultures. Blood 96, 3029–3039 (2000).

    CAS  PubMed  Google Scholar 

  53. Quah, B. J. C., Warren, H. S. & Parish, C. R. Monitoring lymphocyte proliferation in vitro and in vivo with the intracellular fluorescent dye carboxyfluorescein diacetate succinimidyl ester. Nat. Protoc. 2, 2049–2056 (2007).

    CAS  PubMed  Google Scholar 

  54. Li, A. W. et al. A facile approach to enhance antigen response for personalized cancer vaccination. Nat. Mater. 17, 528–534 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Shin, M. D. et al. COVID-19 vaccine development and a potential nanomaterial path forward. Nat. Nanotechnol. 15, 646–655 (2020).

    CAS  PubMed  Google Scholar 

  56. Aldén, M. et al. Intracellular reverse transcription of Pfizer BioNTech COVID-19 mRNA vaccine BNT162b2 in vitro in human liver cell line. Curr. Issues Mol. Biol. 44, 1115–1126 (2022).

    PubMed  PubMed Central  Google Scholar 

  57. Herrmann, I. K., Wood, M. J. A. & Fuhrmann, G. Extracellular vesicles as a next-generation drug delivery platform. Nat. Nanotechnol. 16, 748–759 (2021).

    CAS  PubMed  Google Scholar 

  58. Du, Y. et al. Intranasal administration of a recombinant RBD vaccine induced protective immunity against SARS-CoV-2 in mouse. Vaccine 39, 2280–2287 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Neerukonda, S. N., Vassell, R., Weiss, C. D. & Wang, W. Measuring neutralizing antibodies to SARS-CoV-2 using lentiviral spike-pseudoviruses. Methods Mol. Biol. 2452, 305–314 (2022).

    CAS  PubMed  Google Scholar 

  60. Porrett, P. M. et al. First clinical-grade porcine kidney xenotransplant using a human decedent model. Am. J. Transplant. 22, 1037–1053 (2022).

    PubMed  Google Scholar 

  61. Montgomery, R. A. et al. Results of two cases of pig-to-human kidney xenotransplantation. N. Engl. J. Med. 386, 1889–1898 (2022).

    CAS  PubMed  Google Scholar 

  62. Moazami, N. et al. Pig-to-human heart xenotransplantation in two recently deceased human recipients. Nat. Med. 29, 1989–1997 (2023).

    CAS  PubMed  Google Scholar 

  63. Griffith, B. P. et al. Genetically modified porcine-to-human cardiac xenotransplantation. N. Engl. J. Med. 387, 35–44 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Patience, C., Takeuchi, Y. & Weiss, R. A. Infection of human cells by an endogenous retrovirus of pigs. Nat. Med. 3, 282–286 (1997).

    CAS  PubMed  Google Scholar 

  65. Łopata, K., Wojdas, E., Nowak, R., Łopata, P. & Mazurek, U. Porcine endogenous retrovirus (PERV)—molecular structure and replication strategy in the context of retroviral infection risk of human cells. Front. Microbiol. 9, 730 (2018).

    PubMed  PubMed Central  Google Scholar 

  66. Denner, J. Recent progress in xenotransplantation, with emphasis on virological safety. Ann. Transplant. 21, 717–727 (2016).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Key Research & Development Program of China (grant nos. 2021YFA1201000, 2023YFC2605000, 2024YFA1208301, 2024YFA1212400), National Natural Science Foundation of China (NSFC) (grant nos. 32030060, 82430067, GG9100007028, 22477118). The authors also appreciate the support by the Science Fund for Creative Research Groups of Nature Science Foundation of Hebei Province (B2021201038). This work was also supported by the NSFC (grant no. 82104105), China Postdoctoral Science Foundation 2021M693966. We thank D. Zhang and J. Lian (Core Facility, Center of Biomedical Analysis, Tsinghua University) for technical support with whole LN immunolabeling, tissue clearing, imaging and computational analysis.

Author information

Authors and Affiliations

Authors

Contributions

J.W., N.G. and X.-J.L. conceived and designed the experiments. J.W., N.G., Y.Z., H.X., Y.J., W.L., X.L., F.X. and B.X. performed the experiments. J.W., X.-J.L., Y.Y. and S.S. analysed the results. J.C., Y.Y., X.L., W.G., S.S., J.Z. and A.Z. provided suggestions and technical support. J.L. provided support for the use of light-sheet microscopy for LN imaging and contributed to the discussion and guidance on the characterization of the vaccine. J.W., N.G. and X.-J.L. wrote the manuscript. N.G. and X.-J.L. supervised the entire project. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Ningqiang Gong or Xing-Jie Liang.

Ethics declarations

Competing interests

J.W., N.G. and X.-J.L. have filed a patent application related to this study. The other authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Guangjun Nie and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 IgG modified liposome targets DCs.

a, Illustration of the preparation of liposomes (LIP) and IgG-modified liposomes (LIP-IgG). b, Hydrodynamic diameter and Zeta-potential of LIP and LIP-IgG. c, Quantitative analysis of the uptake of FITC labeled LIP- or LIP-IgG by different cells (3T3, HUVEC, RAW 264.7, DC2.4) using flow cytometry. d, Quantitative analysis of the uptake of LIP, LIP-IgG, or Fc-fragment modified Lip (LIP-Fc) by DC2.4 cells. e-f, DC2.4 cells were pretreated with various concentrations of IgG (e) or Fc fragment (f) and then FITC labeled LIP-IgG were added. After 2 h incubation, mean fluorescence intensity (MFI) of DC2.4 cells were determined using flow cytometry. Data are presented as mean ±s.d., n = 3 independent biological samples per group, statistical differences in c were determined using two-way ANOVA with Tukey’s test for multiple comparison.

Source data

Extended Data Fig. 2 IgG binding to XMVs increases DC uptake.

Mouse subcutaneous tissue homogenate was prepared and centrifuged to remove cells. The supernatant was collected and exploited to treat XMVs (30 min, 37 °C). After that, the XMVs were collected by centrifugation and washed with PBS. a, A representative TEM image of XMV after stained with an anti-IgG antibody-conjugated to gold bead. Scare bar, 50 nm. Liquid chromatography tandem mass spectrometry (LC-MS/MS) was used to identify and quantify the components of the protein corona. Classifications of components of the proteins (b) and their isoelectric point (pI) (c) of the top 21 abundant proteins. d, Gating strategies for analyzing antibody binding to XMVs. e, The binding of IgG, IgA, or IgM to XMVs after XMVs were incubated with subcutaneous tissue homogenate, analyzed using a CytoFLEX nanoflow cytometry. f, XMVs from different cell lines (PED, Vero) were used to prepare XMV-Ag. Mouse IgG was mixed with XMV-Ag for 30 min to form IgG-XMV-Ag. Various XMV-Ag, AUV-Ag (AUVs were derived from mouse RBC cells), or Lip- Ag were used to treat DC2.4 cells. After 2 h, The uptake of the antigen peptide by DC2.4 were determined using flow cytometry. g, XMV-Ag were preincubated with different amounts of IgG and then were added to DC2.4 cells. After 2 h, MFI of DC2.4 cells were determined using flow cytometry. h, XMV-Ag or XMV-Ag with IgG-preincubation were used to treat DC2.4 cells in the absence or present of FcR blockade. i, BMDCs were incubated with the XMV-Ag for 24 h and the presentation of SIINFEKL peptide on DC surface were stained using a H-2 K(b) OVA (SIINFEKL) antibody. After that, the cells were observed using a confocal microscopy. Green, Ag/MHC-I; blue, nuclei. Data are presented as mean ±s.d. and statistical differences in f were determined using one-way ANOVA with Tukey’s test for multiple comparison.

Source data

Extended Data Fig. 3 XMV-Ag targets DCs and promotes DC maturation in vivo.

Mice were subcutaneously injected with FITC labeled XMV-Ag at 0 h. After 24 h, the LNs were collected and the percentages of cellular uptake of FITC-XMV-Ag in LNs were determined using flow cytometry. a. Flow cytometry analysis of cellular uptake of XMV-Ag in LNs. Representative flow dot plots (left) and the percentages of XMV-Ag uptake by various cells in LNs (right) are shown. b. Gating strategy for flow cytometry analysis of DC subtypes such as cDC1(CD317-B220-CD11c+MHC-II+XCR1+), cDC2(CD317-B220-CD11c+MHC-II+CD172+) and pDC (CD317+B220+) in LNs. Mice were treated with XMV-Ag for 24 h and the percentages of CD11c+ CD40+ DCs (c) and CD11c+ CD86+ DCs (d) in LNs were determined using flow cytometry. Data are presented as mean ± s.d. Data in a, c-d, n = 3 independent biological samples per group.

Source data

Extended Data Fig. 4 XMV-Ag induces T-cell immune responses.

a, Flow cytometry analysis of OVA tetramer+CD8+ T cells in total blood immune cells. b, Percentages of IFN-γ+ cells in splenic CD8+CD44+ T cells from mice treated with three vaccinations of XMV-Ag, AUV-Ag, LIP-Ag, free Ag, or PBS. Representative flow dot plots (c-d) and quantifications (e-f) of memory cells in CD4+ or CD8+ T cells, naive T cells in CD4+ or CD8+ T cells in spleen cells after three-doses of XMV-Ag, AUV-Ag, LIP-Ag, free Ag, or PBS. Data are presented as mean ±s.d.(n = 4), statistical differences were determined using one-way ANOVA with Tukey’s test for multiple comparison.

Source data

Supplementary information

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

Wang, J., Zhang, Y., Jia, Y. et al. Targeting vaccines to dendritic cells by mimicking the processing and presentation of antigens in xenotransplant rejection. Nat. Biomed. Eng 9, 201–214 (2025). https://doi.org/10.1038/s41551-025-01343-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41551-025-01343-6

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