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:

Synthetically glycosylated antigens for the antigen-specific suppression of established immune responses

Abstract

Inducing antigen-specific tolerance during an established immune response typically requires non-specific immunosuppressive signalling molecules. Hence, standard treatments for autoimmunity trigger global immunosuppression. Here we show that established antigen-specific responses in effector T cells and memory T cells can be suppressed by a polymer glycosylated with N-acetylgalactosamine (pGal) and conjugated to the antigen via a self-immolative linker that allows for the dissociation of the antigen on endocytosis and its presentation in the immunoregulatory environment. We show that pGal–antigen therapy induces antigen-specific tolerance in a mouse model of experimental autoimmune encephalomyelitis (with programmed cell-death-1 and the co-inhibitory ligand CD276 driving the tolerogenic responses), as well as the suppression of antigen-specific responses to vaccination against a DNA-based simian immunodeficiency virus in non-human primates. Our findings show that pGal–antigen therapy invokes mechanisms of immune tolerance to resolve antigen-specific inflammatory T-cell responses and suggest that the therapy may be applicable across autoimmune diseases.

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: Antigen-mediated suppression of CFA/IFA recall responses in transgenic T cells.
Fig. 2: Antigen-mediated suppression of R848 recall responses in transgenic T cells.
Fig. 3: Transcriptional profile of the induction of therapeutic T-cell tolerance.
Fig. 4: Effects of PD-1 blockade on the antigen-mediated suppression of CFA/IFA recall responses in transgenic T cells.
Fig. 5: Therapeutic suppression of the responses to adoptive transfer in a model of chronic EAE.
Fig. 6: Therapeutic suppression of relapsing-remitting EAE.
Fig. 7: Antigen-specific suppression of T-cell immunity with pGal antigen therapy in NHPs.

Similar content being viewed by others

Data availability

Gene-expression data obtained by RNA-seq are available from the NCBI Gene Expression Omnibus, via the accession number GSE192671. Source data are provided with this paper. All other data needed to evaluate the conclusions of the study are available within the paper and its Supplementary Information.

References

  1. Kapp, K. et al. Modulation of systemic antigen-specific immune responses by oral antigen in humans. Eur. J. Immunol. 40, 3128–3137 (2010).

    Article  CAS  PubMed  Google Scholar 

  2. Peng, H. J., Turner, M. W. & Strobel, S. The generation of a ‘tolerogen’ after the ingestion of ovalbumin is time-dependent and unrelated to serum levels of immunoreactive antigen. Clin. Exp. Immunol. 81, 510–515 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kawamoto, S. et al. Foxp3+ T cells regulate immunoglobulin A selection and facilitate diversification of bacterial species responsible for immune homeostasis. Immunity 41, 152–165 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. Rouhani, S. J. et al. Roles of lymphatic endothelial cells expressing peripheral tissue antigens in CD4 T-cell tolerance induction. Nat. Commun. 6, 6771 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Kurts, C., Kosaka, H., Carbone, F. R., Miller, J. F. A. P. & Heath, W. R. Class I-restricted cross-presentation of exogenous self-antigens leads to deletion of autoreactive CD8+ T cells. J. Exp. Med. 186, 239–245 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Pape, K. A., Merica, R., Mondino, A., Khoruts, A. & Jenkins, M. K. Direct evidence that functionally impaired CD4+ T cells persist in vivo following induction of peripheral tolerance. J. Immunol. 160, 4719–4729 (1998).

    Article  CAS  PubMed  Google Scholar 

  7. Martinez, R. J. et al. Arthritogenic self-reactive CD4+ T cells acquire an FR4hiCD73hi anergic state in the presence of Foxp3+ regulatory T cells. J. Immunol. 188, 170–181 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Angelosanto, J. M., Blackburn, S. D., Crawford, A. & Wherry, E. J. Progressive loss of memory T cell potential and commitment to exhaustion during chronic viral infection. J. Virol. 86, 8161–8170 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Takahashi, T. et al. Immunologic self-tolerance maintained by CD25+CD4+ naturally anergic and suppressive T cells: induction of autoimmune disease by breaking their anergic/suppressive state. Int. Immunol. 10, 1969–1980 (1998).

    Article  CAS  PubMed  Google Scholar 

  10. Hori, S., Nomura, T. & Sakaguchi, S. Control of regulatory T cell development by the transcription factor Foxp3. J. Immunol. 198, 981–985 (2017).

    CAS  PubMed  Google Scholar 

  11. Wilson, D. S. et al. Synthetically glycosylated antigens induce antigen-specific tolerance and prevent the onset of diabetes. Nat. Biomed. Eng. 3, 817–829 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Kreuwel, H. T. C., Aung, S., Silao, C. & Sherman, L. A. Memory CD8+ T cells undergo peripheral tolerance. Immunity 17, 73–81 (2002).

    Article  CAS  PubMed  Google Scholar 

  13. Edwards, J. C. W. & Cambridge, G. Sustained improvement in rheumatoid arthritis following a protocol designed to deplete B lymphocytes. Rheumatology 40, 205–211 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Leung, D. Y. M. et al. Effect of anti-IgE therapy in patients with peanut allergy. N. Engl. J. Med. 348, 986–993 (2003).

    Article  CAS  PubMed  Google Scholar 

  15. Azzi, J. R., Sayegh, M. H. & Mallat, S. G. Calcineurin inhibitors: 40 years later, can’t live without …. J. Immunol. 191, 5785–5791 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Nousari, H. C., Sragovich, A., Kimyai-Asadi, A., Orlinsky, D. & Anhalt, G. J. Mycophenolate mofetil in autoimmune and inflammatory skin disorders. J. Am. Acad. Dermatol. 40, 265–268 (1999).

    Article  CAS  PubMed  Google Scholar 

  17. Chatrath, H., Allen, L. & Boyer, T. D. Use of sirolimus in the treatment of refractory autoimmune hepatitis. Am. J. Med. 127, 1128–1131 (2014).

    Article  CAS  PubMed  Google Scholar 

  18. Godeau, B. et al. Intravenous immunoglobulin or high-dose methylprednisolone, with or without oral prednisone, for adults with untreated severe autoimmune thrombocytopenic purpura: a randomised, multicentre trial. Lancet 359, 23–29 (2002).

    Article  CAS  PubMed  Google Scholar 

  19. Kempen, J. H. et al. Long-term risk of malignancy among patients treated with immunosuppressive agents for ocular inflammation: a critical assessment of the evidence. Am. J. Ophthalmol. 146, 802–812 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Casella, G. et al. Oligodendrocyte-derived extracellular vesicles as antigen-specific therapy for autoimmune neuroinflammation in mice. Sci. Transl. Med. 12, eaba0599 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Krienke, C. et al. A noninflammatory mRNA vaccine for treatment of experimental autoimmune encephalomyelitis. Science 371, 145–153 (2021).

    Article  CAS  PubMed  Google Scholar 

  22. Maulloo, C. D. et al. Lymph node-targeted synthetically glycosylated antigen leads to antigen-specific immunological tolerance. Front. Immunol. 12, 1–17 (2021).

    Article  Google Scholar 

  23. Damo, M., Wilson, D. S., Watkins, E. A. & Hubbell, J. A. Soluble N-acetylgalactosamine-modified antigens enhance hepatocyte-dependent antigen cross-presentation and result in antigen-specific CD8+ T cell tolerance development. Front. Immunol. 12, 1–15 (2021).

    Article  Google Scholar 

  24. Horst, A. K., Neumann, K., Diehl, L. & Tiegs, G. Modulation of liver tolerance by conventional and nonconventional antigen-presenting cells and regulatory immune cells. Cell. Mol. Immunol. 13, 277–292 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Li, F. & Tian, Z. The liver works as a school to educate regulatory immune cells. Cell. Mol. Immunol. 10, 292–302 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Stoop, J. N., Tibbitt, C. A., van Eden, W., Robinson, J. H. & Hilkens, C. M. U. The choice of adjuvant determines the cytokine profile of T cells in proteoglycan-induced arthritis but does not influence disease severity. Immunology 138, 68–75 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Khan, O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kalekar, L. A. et al. CD4+ T cell anergy prevents autoimmunity and generates regulatory T cell precursors. Nat. Immunol. 17, 304–314 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hataye, J., Moon, J. J., Khoruts, A., Reilly, C. & Jenkins, M. K. Naïve and memory CD4. Science 312, 114–116 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. Marzo, A. L. et al. Initial T cell frequency dictates memory CD8+ T cell lineage commitment. Nat. Immunol. 6, 793–799 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Bautista, J. L. et al. Intraclonal competition limits the fate determination of regulatory T cells in the thymus. Nat. Immunol. 10, 610–617 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Gustafson, C. E. et al. Systems analysis of microRNAs reveals a novel link between CD8 T cell dysfunction and TGF-β signaling in immune aging. J. Immunol. 196, 54-14 (2016).

    Article  Google Scholar 

  33. Raman, C. et al. TGF-β receptor 3 (betaglycan) regulates Th1 differentiation and T dependent B cell responses. J. Immunol 196, 189.12 (2016).

    Article  Google Scholar 

  34. Szabo, S. J. et al. Distinct effects of T-bet in Th1 lineage commitment and IFN-γ production in CD4 and CD8 T cells. Science 295, 338–342 (2002).

    Article  CAS  PubMed  Google Scholar 

  35. Shaw, L. A. et al. Id2 reinforces TH 1 differentiation and inhibits E2A to repress TFH differentiation. Nat. Immunol. 17, 834–843 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Murga, M. et al. Mutation of E2F2 in mice causes enhanced T lymphocyte proliferation, leading to the development of autoimmunity. Immunity 15, 959–970 (2001).

    Article  CAS  PubMed  Google Scholar 

  37. Watanabe, M., Moon, K. D., Vacchio, M. S., Hathcock, K. S. & Hodes, R. J. Downmodulation of tumor suppressor p53 by T cell receptor signaling is critical for antigen-specific CD4+ T cell responses. Immunity 40, 681–691 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Barili, V. et al. Targeting p53 and histone methyltransferases restores exhausted CD8+ T cells in HCV infection. Nat. Commun. 11, 604 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Banerjee, A. et al. Lack of p53 augments antitumor functions in cytolytic T Cells. Cancer Res. 76, 5229–5240 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Fang, F. et al. Human transcription factor KLF3 maintains T lymphocyte quiescent phenotype via inhibiting SHP-1 expression. Blood 126, 3426 (2015).

    Article  Google Scholar 

  41. Escobar, G., Mangani, D. & Anderson, A. C. T cell factor 1: a master regulator of the T cell response in disease. Sci. Immunol. 5, eabb9726 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Rauch, K. S. et al. Id3 maintains Foxp3 expression in regulatory T cells by controlling a transcriptional network of E47, Spi-B, and SOCS3. Cell Rep. 17, 2827–2836 (2016).

    Article  CAS  PubMed  Google Scholar 

  43. Karpuzoglu, E., Phillips, R. A., Gogal, R. M. & Ansar Ahmed, S. IFN-gamma-inducing transcription factor, T-bet is upregulated by estrogen in murine splenocytes: role of IL-27 but not IL-12. Mol. Immunol. 44, 1808–1814 (2007).

    Article  CAS  PubMed  Google Scholar 

  44. Maret, A. et al. Estradiol enhances primary antigen-specific CD4 T cell responses and Th1 development in vivo. Essential role of estrogen receptor alpha expression in hematopoietic cells. Eur. J. Immunol. 33, 512–521 (2003).

    Article  CAS  PubMed  Google Scholar 

  45. von Knethen, A. et al. Tolerizing CTL by sustained hepatic PD-L1 expression provides a new therapy approach in mouse sepsis. Theranostics 9, 2003–2016 (2019).

    Article  Google Scholar 

  46. Hutchins, N. A., Wang, F., Wang, Y., Chung, C.-S. & Ayala, A. Kupffer cells potentiate liver sinusoidal endothelial cell injury in sepsis by ligating programmed cell death ligand-1. J. Leukoc. Biol. 94, 963–970 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Iwai, Y., Terawaki, S., Ikegawa, M., Okazaki, T. & Honjo, T. PD-1 inhibits antiviral immunity at the effector phase in the liver. J. Exp. Med. 198, 39–50 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Dong, H. et al. B7-H1 determines accumulation and deletion of intrahepatic CD8+ T lymphocytes. Immunity 20, 327–336 (2004).

    Article  CAS  PubMed  Google Scholar 

  49. Martinon, F. et al. Persistent immune responses induced by a human immunodeficiency virus DNA vaccine delivered in association with electroporation in the skin of nonhuman primates. Hum. Gene Ther. 20, 1291–1307 (2009).

    Article  CAS  PubMed  Google Scholar 

  50. Adam, L. et al. Innate molecular and cellular signature in the skin preceding long-lasting T cell responses after electroporated DNA vaccination. J. Immunol. 204, 3375–3388 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Anderson, A. C., Joller, N. & Kuchroo, V. K. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity 44, 989–1004 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Dulos, J. et al. PD-1 blockade augments Th1 and Th17 and suppresses Th2 responses in peripheral blood from patients with prostate and advanced melanoma cancer. J. Immunother. 35, 169–178 (2012).

    Article  CAS  PubMed  Google Scholar 

  53. Wang, S. C. et al. Programmed cell death-1 (PD-1) and T-cell immunoglobulin mucin-3 (Tim-3) regulate CD4+ T cells to induce type 2 helper T cell (Th2) bias at the maternal–fetal interface. Hum. Reprod. 31, 700–711 (2016).

    Article  CAS  PubMed  Google Scholar 

  54. Joller, N. et al. Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses. Immunity 40, 569–581 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Cameron, M. J. et al. IL-4 prevents insulitis and insulin-dependent diabetes mellitus in nonobese diabetic mice by potentiation of regulatory T helper-2 cell function. J. Immunol. 159, 4686–4692 (1997).

    Article  CAS  PubMed  Google Scholar 

  56. Racke, M. K. et al. Cytokine-induced immune deviation as a therapy for inflammatory autoimmune disease. J. Exp. Med. 180, 1961–1966 (1994).

    Article  CAS  PubMed  Google Scholar 

  57. Murray, J. A. et al. Safety and tolerability of KAN-101, a liver-targeted immune tolerance therapy, in patients with coeliac disease (ACeD): a phase 1 trial. Lancet Gastroenterol. Hepatol. https://doi.org/10.1016/S2468-1253(23)00107-3 (2023).

    Article  PubMed  Google Scholar 

  58. Roederer, M., Nozzi, J. L. & Nason, M. C. SPICE: exploration and analysis of post-cytometric complex multivariate datasets. Cytometry A 79A, 167–174 (2011).

    Article  Google Scholar 

Download references

Acknowledgements

We thank B. Jabri and P. A. Savage, both of the University of Chicago, for helpful discussions. We thank the Cytometry and Antibody Core Facility at the University of Chicago. We thank Hooke Laboratories for adoptive transfer EAE studies. We thank the Center for Research Informatics, which is funded by the Biological Sciences Division at the University of Chicago with additional funding from the Institute for Translational Medicine, CTSA grant number UL1 TR000430 from the NIH, and the University of Chicago Comprehensive Cancer Center Support Grant (NIH P30CA014599). Tetramers were provided by the NIH Tetramer Core Facility. We thank B. Delache, S. Langois, O. Lacroix, N. Dhooge, J.M. Robert, T. Prot and C. Dodan for the NHP experiments; L. Bossevot, M. Leonec, A. Chatenet, J. Morin and M. Gomez-Pacheco for the NHP ELISPOT assays and flow cytometry; S. Gomes for assistance with experiments; and L. Shores for assistance with manuscript editing. This work was supported by the University of Chicago’s Chicago Immunoengineering Innovation Center, the Alper Family Foundation and Anokion S.A. The Infectious Disease Models and Innovative Therapies research infrastructure is supported by the ‘Programme Investissements d’Avenir’, managed by the Agence Nationale de la Recherche (ANR) (ANR-11-INBS-0008 and ANR-10-EQPX-02-01).

Author information

Authors and Affiliations

Authors

Contributions

A.C.T., K.M.L., R.L., N.D.-B., S.K. and J.A.H. designed the research strategy. A.C.T., R.P.W., T.B.T., J.T.A., A.T.A., E.A.W., C.D.M., E.B., J.W.R., A.S., M.N., R. Marlin., N.D.-B. and A.-S.G. performed the experiments. A.C.T., J.T.A., R.P.W., J.W.R., A.T.A., R. Mishra., N.D.-B., A.-S.G., T.B.T., R. Marlin. and K.M.L. analysed the experiments. D.S.W., S.K., G.P.C., K.M.L. and J.A.H. conceptualized the materials. D.S.W., D.J.B., J.L.H., M.R.R. and A.J.S. synthesized the materials. A.C.T., R.P.W., S.K. and J.A.H. wrote the manuscript.

Corresponding authors

Correspondence to D. Scott Wilson or Jeffrey A. Hubbell.

Ethics declarations

Competing interests

This work was funded in part by Anokion SA. D.S.W., J.A.H., S.K. and K.M.L. are inventors on patents related to synthetically glycosylated antigens, licensed to Anokion SA. J.A.H. consults for Anokion, is on the Board of Directors of the company and on its Scientific Advisory Board, and holds equity in the company. T.B.T., D.J.B., J.L.H., R.M., and G.P.C. are employees of Anokion. S.K. is on the Scientific Advisory Board of Anokion and holds equity in the company. K.M.L. also holds equity in Anokion. All other authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks the anonymous reviewers 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 Conjugation of pGal to the antigen is required for tolerance induction.

OTI and OTII cells were adoptively transferred into mice prior to vaccination with CFA/OVA as in Fig. 1. 4 weeks later mice were treated with either pGal alone, an unconjugated mix of pGal and OVA or saline prior to challenge. (a) Percent recovery of OTI and OTII cells in dLN and spleens as a proportion of the CD8+ and CD4+ T-cell populations, respectively. (b) Expression frequencies of the specified protein markers as indicated on the y-axes on recovered OTI and OII cells from SLO. (c) Three-day restimulations were also carried out for ELISA-based analyses of the indicated cytokines in culture supernatants. Data are shown as means ± SEM. Unless otherwise stated statistical differences in all graphs were determined by one-way ANOVA with Tukey’s test.

Source data

Extended Data Fig. 2 Six-hour restimulation data, from Fig. 1.

a–d) As in Fig. 1, six-hour ex vivo antigen restimulations were performed with recovered lymphocytes and peptides as indicated above each graph, and intracellular flow cytometry was used to detect production of the indicated cytokines in OTI and OTII cells. Data are shown as means ± SEM. Each point represents an individual mouse. Unless otherwise stated statistical differences in all graphs were determined by one-way ANOVA with Tukey’s test.

Source data

Extended Data Fig. 3 Six-hour restimulation data, from Fig. 2.

As in Fig. 2, six-hour ex vivo antigen restimulations were performed with recovered lymphocytes and peptides as indicated above each graph, and intracellular flow cytometry was used to detect production of the indicated cytokines in OTI and OTII cells. Data are shown as means ± SEM. Each point represents an individual mouse. Unless otherwise stated statistical differences in all graphs were determined by one-way ANOVA with Tukey’s test.

Source data

Extended Data Fig. 4 pGal-OVA R848 recall response dosing.

(a–d) MFI of the indicated markers on the y-axes in recovered OTI&II cells. (e, f) Expression frequencies of the specified protein markers as indicated on the y-axes on recovered OTIs from SLO. Data are shown as means ± SEM. Unless otherwise stated statistical differences in all graphs were determined by one-way ANOVA with Tukey’s test.

Source data

Extended Data Fig. 5 Antigen-mediated suppression of LmOVA/CFA recall response in endogenous CD8+ T cells.

Mice were first inoculated with LmOVA and allowed to clear the infection, then one month later given antigen therapy as described previously, and finally challenged two weeks after the last antigen dose with OVA+CFA s.c. a) Quantification of endogenous OVA-specific CD8+T cells recovered in dLN and spleens as a proportion of total CD8+ T cells as well as total cell number. b) Representative flow cytometry profiles of recovered pentamer-reactive CD8+ T cells in the dLN comparing effects of saline and antigen treatments. c–f) Expression frequencies of the specified protein markers as indicated on the y-axes on recovered pentamer-specific CD8+ T cells from SLO. g–i) Six-hour ex vivo antigen restimulations were performed with recovered lymphocytes and peptides as indicated above each graph, and intracellular flow cytometry was used to detect production of the indicated cytokines in pentamer-reactive CD8+ T cells. *p indicates comparisons between pGal and Saline groups, &p indicates comparisons between OVA and Saline groups, #p indicates comparisons between pGal and OVA groups. Data are shown as means ± SEM. Unless otherwise stated statistical differences in all graphs were determined by one-way ANOVA with Tukey’s post hoc test.

Source data

Extended Data Fig. 6 Six-hour restimulation data, from Fig. 4.

a–c) As in Fig. 4, six-hour ex vivo antigen restimulations were performed with recovered lymphocytes and peptides as indicated above each graph, and intracellular flow cytometry was used to detect production of the indicated cytokines in OTI and OTII cells. Data are shown as means ± SEM. Each point represents an individual mouse. Unless otherwise stated statistical differences in all graphs were determined by one-way ANOVA with Tukey’s test.

Source data

Extended Data Fig. 7 Relapsing-remitting EAE peptide restimulation.

(a) Six-hour ex vivo antigen restimulations were performed as indicated and intracellular flow cytometry was used to detect production of the indicated cytokines in CD8+and CD4+T cells. (b) Three-day restimulations were carried out for analysis of IL-17A production in culture supernatants. All statistical comparisons were performed by one-way ANOVA using Tukey correction unless otherwise indicated.

Source data

Supplementary information

Supplementary Information

Supplementary methods, figures, tables and references.

Reporting Summary

Peer Review File

Supplementary Data

Raw data for the supplementary figures.

Supplementary Table

Supplementary Table 16.

Supplementary Table

Supplementary Tables 1–13.

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

Tremain, A.C., Wallace, R.P., Lorentz, K.M. et al. Synthetically glycosylated antigens for the antigen-specific suppression of established immune responses. Nat. Biomed. Eng 7, 1142–1155 (2023). https://doi.org/10.1038/s41551-023-01086-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-023-01086-2

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