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:

mRNA vaccine against malaria tailored for liver-resident memory T cells

Abstract

Malaria is caused by Plasmodium species transmitted by Anopheles mosquitoes. Following a mosquito bite, Plasmodium sporozoites migrate from skin to liver, where extensive replication occurs, emerging later as merozoites that can infect red blood cells and cause symptoms of disease. As liver tissue-resident memory T cells (Trm cells) have recently been shown to control liver-stage infections, we embarked on a messenger RNA (mRNA)-based vaccine strategy to induce liver Trm cells to prevent malaria. Although a standard mRNA vaccine was unable to generate liver Trm or protect against challenge with Plasmodium berghei sporozoites in mice, addition of an agonist that recruits T cell help from type I natural killer T cells under mRNA-vaccination conditions resulted in significant generation of liver Trm cells and effective protection. Moreover, whereas previous exposure of mice to blood-stage infection impaired traditional vaccines based on attenuated sporozoites, mRNA vaccination was unaffected, underlining the potential for such a rational mRNA-based strategy in malaria-endemic regions.

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: Adjuvanted mRNA vaccines can induce liver Trm cells and protect against sporozoite challenge.
Fig. 2: Defining the ideal adjuvant for liver Trm cell induction.
Fig. 3: Dissection of the mechanistic basis for vaccine activity.
Fig. 4: Endogenous T cell response to adjuvanted mRNA vaccination.
Fig. 5: Vaccination with adjuvanted mRNA can induce protection against an authentic parasite antigen.
Fig. 6: Protective responses can be boosted and require CD8 cells.
Fig. 7: Protection is largely dependent on liver Trm cells.
Fig. 8: Vaccination with adjuvanted mRNA is not impaired by preexposure to blood-stage malaria.

Similar content being viewed by others

Data availability

Data to support the findings of study are available from the corresponding author upon request without restrictions. Source data are provided with this paper.

References

  1. World Health Organization. World Malaria Report 2021. WHO, 2021.

  2. Cowman, A. F., Healer, J., Marapana, D. & Marsh, K. Malaria: biology and disease. Cell 167, 610–624 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Crompton, P. D. et al. Malaria immunity in man and mosquito: insights into unsolved mysteries of a deadly infectious disease. Annu Rev. Immunol. 32, 157–187 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Laurens, M. B. RTS,S/AS01 vaccine (Mosquirix): an overview. Hum. Vaccin Immunother. 16, 480–489 (2020).

    Article  CAS  PubMed  Google Scholar 

  5. Duffy, P. E. & Patrick Gorres, J. Malaria vaccines since 2000: progress, priorities, products. NPJ Vaccines 5, 48 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Datoo, M. S. et al. Efficacy and immunogenicity of R21/Matrix-M vaccine against clinical malaria after 2 years’ follow-up in children in Burkina Faso: a phase 1/2b randomised controlled trial. Lancet Infect. Dis. 22, 1728–1736 (2022).

    Article  CAS  PubMed  Google Scholar 

  7. Datoo, M. S. et al. Efficacy of a low-dose candidate malaria vaccine, R21 in adjuvant Matrix-M, with seasonal administration to children in Burkina Faso: a randomised controlled trial. Lancet 397, 1809–1818 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Fernandez-Ruiz, D. et al. Liver-resident memory CD8+ T cells form a front-line defense against malaria liver-stage infection. Immunity 45, 889–902 (2016).

    Article  CAS  PubMed  Google Scholar 

  9. Seder, R. A. et al. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science 341, 1359–1365 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. Epstein, J. E. et al. Live attenuated malaria vaccine designed to protect through hepatic CD8+ T cell immunity. Science 334, 475–480 (2011).

    Article  CAS  PubMed  Google Scholar 

  11. Schofield, L. et al. γ Interferon, CD8+ T cells and antibodies required for immunity to malaria sporozoites. Nature 220, 664–666 (1987).

    Article  Google Scholar 

  12. Lefebvre, M. N. et al. Expeditious recruitment of circulating memory CD8 T cells to the liver facilitates control of malaria. Cell Rep. 37, 109956 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Tse, S. W., Radtke, A. J., Espinosa, D. A., Cockburn, I. A. & Zavala, F. The chemokine receptor CXCR6 is required for the maintenance of liver memory CD8+ T cells specific for infectious pathogens. J. Infect. Dis. 210, 1508–1516 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Holz, L. E. et al. CD8+ T cell activation leads to constitutive formation of liver tissue-resident memory T cells that seed a large and flexible niche in the liver. Cell Rep. 25, 68–79 (2018).

    Article  CAS  PubMed  Google Scholar 

  15. Nunes-Cabaco, H., Moita, D. & Prudencio, M. Five decades of clinical assessment of whole-sporozoite malaria vaccines. Front. Immunol. 13, 977472 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ishizuka, A. S. et al. Protection against malaria at 1 year and immune correlates following PfSPZ vaccination. Nat. Med. 22, 614–623 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Jongo, S. A. et al. Immunogenicity and protective efficacy of radiation-attenuated and chemo-attenuated PfSPZ vaccines in Equatoguinean adults. Am. J. Trop. Med. Hyg. 104, 283–293 (2021).

    Article  CAS  PubMed  Google Scholar 

  18. Oneko, M. et al. Safety, immunogenicity and efficacy of PfSPZ vaccine against malaria in infants in western Kenya: a double-blind, randomized, placebo-controlled phase 2 trial. Nat. Med. 27, 1636–1645 (2021).

    Article  CAS  PubMed  Google Scholar 

  19. Sissoko, M. S. et al. Safety and efficacy of a three-dose regimen of Plasmodium falciparum sporozoite vaccine in adults during an intense malaria transmission season in Mali: a randomised, controlled phase 1 trial. Lancet Infect. Dis. 22, 377–389 (2022).

    Article  CAS  PubMed  Google Scholar 

  20. Ocana-Morgner, C., Mota, M. M. & Rodriguez, A. Malaria blood stage suppression of liver stage immunity by dendritic cells. J. Exp. Med. 197, 143–151 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Olsen, T. M., Stone, B. C., Chuenchob, V. & Murphy, S. C. Prime-and-trap malaria vaccination to generate protective CD8+ liver-resident memory T cells. J. Immunol. 201, 1984–1993 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Gola, A. et al. Prime and target immunization protects against liver-stage malaria in mice. Sci. Transl. Med. 10, eaap9128 (2018).

    Article  PubMed  Google Scholar 

  23. Holz, L. E. et al. Glycolipid-peptide vaccination induces liver-resident memory CD8+ T cells that protect against rodent malaria. Sci. Immunol. 5, eaaz8035 (2020).

    Article  CAS  PubMed  Google Scholar 

  24. Brennan, P. J., Brigl, M. & Brenner, M. B. Invariant natural killer T cells: an innate activation scheme linked to diverse effector functions. Nat. Rev. Immunol. 13, 101–117 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. Heide, J., Vaughan, K. C., Sette, A., Jacobs, T. & Schulze Zur Wiesch, J. Comprehensive Review of human Plasmodium falciparum-specific CD8+ T cell epitopes. Front. Immunol. 10, 397 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Morita, M. et al. Structure-activity relationship of alpha-galactosylceramides against B16-bearing mice. J. Med. Chem. 38, 2176–2187 (1995).

    Article  CAS  PubMed  Google Scholar 

  27. Hogquist, K. A. et al. T cell receptor antagonist peptides induce positive selection. Cell 76, 17–27 (1994).

    Article  CAS  PubMed  Google Scholar 

  28. Compton, B. J. et al. Enhancing T cell responses and tumour immunity by vaccination with peptides conjugated to a weak NKT cell agonist. Org. Biomol. Chem. 17, 1225–1237 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Compton, B. J. et al. Synthesis and activity of 6′′-deoxy-6′′-thio-α-GalCer and peptide conjugates. Org. Lett. 17, 5954–5957 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Hung, J. T. et al. Design and synthesis of galactose-6-OH-modified α-galactosyl ceramide analogues with Th2-biased immune responses. RSC Adv. 4, 47341–47356 (2014).

    Article  CAS  Google Scholar 

  31. Chennamadhavuni, D. et al. Dual modifications of α-galactosylceramide synergize to promote activation of human invariant natural killer T cells and stimulate anti-tumor immunity. Cell Chem. Biol. 25, 571–584 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Christo, S. N. et al. Discrete tissue microenvironments instruct diversity in resident memory T cell function and plasticity. Nat. Immunol. 22, 1140–1151 (2021).

    Article  CAS  PubMed  Google Scholar 

  33. Carvalho, L. H. et al. IL-4-secreting CD4+ T cells are crucial to the development of CD8+ T-cell responses against malaria liver stages. Nat. Med. 8, 166–170 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Valencia-Hernandez, A. M. et al. A natural peptide antigen within the Plasmodium ribosomal protein RPL6 confers liver TRM cell-mediated immunity against malaria in mice. Cell Host Microbe 27, 950–962 (2020).

    Article  CAS  PubMed  Google Scholar 

  35. Evrard, M. et al. Sphingosine 1-phosphate receptor 5 (S1PR5) regulates the peripheral retention of tissue-resident lymphocytes. J. Exp. Med. 219, e20210116 (2022).

    Article  CAS  PubMed  Google Scholar 

  36. Grunwitz, C. et al. HPV16 RNA-LPX vaccine mediates complete regression of aggressively growing HPV-positive mouse tumors and establishes protective T cell memory. Oncoimmunology 8, e1629259 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Salomon, N. et al. Local radiotherapy and E7 RNA-LPX vaccination show enhanced therapeutic efficacy in preclinical models of HPV16+ cancer. Cancer Immunol. Immunother. 71, 1975–1988 (2022).

    Article  CAS  PubMed  Google Scholar 

  38. Salomon, N. et al. A liposomal RNA vaccine inducing neoantigen-specific CD4+ T cells augments the antitumor activity of local radiotherapy in mice. Oncoimmunology 9, 1771925 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Sahin, U. et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature 585, 107–112 (2020).

    Article  CAS  PubMed  Google Scholar 

  40. Kranz, L. M. et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 534, 396–401 (2016).

    Article  PubMed  Google Scholar 

  41. Mallory, K. L. et al. Messenger RNA expressing PfCSP induces functional, protective immune responses against malaria in mice. NPJ Vaccines 6, 84 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Godfrey, D. I., Le Nours, J., Andrews, D. M., Uldrich, A. P. & Rossjohn, J. Unconventional T cell targets for cancer immunotherapy. Immunity 48, 453–473 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Giaccone, G. et al. A phase I study of the natural killer T-cell ligand alpha-galactosylceramide (KRN7000) in patients with solid tumors. Clin. Cancer Res. 8, 3702–3709 (2002).

    CAS  PubMed  Google Scholar 

  44. Tefit, J. N. et al. Efficacy of ABX196, a new NKT agonist, in prophylactic human vaccination. Vaccine 32, 6138–6145 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Guevara, M. L., Jilesen, Z., Stojdl, D. & Persano, S. Codelivery of mRNA with α-galactosylceramide using a new lipopolyplex formulation induces a strong antitumor response upon intravenous administration. ACS Omega 4, 13015–13026 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Verbeke, R. et al. Broadening the message: a nanovaccine co-loaded with messenger RNA and α-GalCer induces antitumor immunity through conventional and natural killer T cells. ACS Nano 13, 1655–1669 (2019).

    CAS  PubMed  Google Scholar 

  47. Miyamoto, K., Miyake, S. & Yamamura, T. A synthetic glycolipid prevents autoimmune encephalomyelitis by inducing TH2 bias of natural killer T cells. Nature 413, 531–534 (2001).

    Article  CAS  PubMed  Google Scholar 

  48. Kain, L. et al. The identification of the endogenous ligands of natural killer T cells reveals the presence of mammalian α-linked glycosylceramides. Immunity 41, 543–554 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Schmidt, N. W. et al. Memory CD8 T cell responses exceeding a large but definable threshold provide long-term immunity to malaria. Proc. Natl Acad. Sci. USA 105, 14017–14022 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Murphy, S. C., Kas, A., Stone, B. C. & Bevan, M. J. A T-cell response to a liver-stage Plasmodium antigen is not boosted by repeated sporozoite immunizations. Proc. Natl Acad. Sci. USA 110, 6055–6060 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Wilson, N. S. et al. Systemic activation of dendritic cells by Toll-like receptor ligands or malaria infection impairs cross-presentation and antiviral immunity. Nat. Immunol. 7, 165–172 (2006).

    Article  CAS  PubMed  Google Scholar 

  52. Chandra, S. et al. A new mouse strain for the analysis of invariant NKT cell function. Nat. Immunol. 16, 799–800 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kawabe, T. et al. The immune responses in CD40-deficient mice: impaired immunoglobulin class switching and germinal center formation. Immunity 1, 167–178 (1994).

    Article  CAS  PubMed  Google Scholar 

  54. Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science 322, 1097–1100 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kennedy, M. K. et al. Reversible defects in natural killer and memory CD8 T cell lineages in interleukin 15-deficient mice. J. Exp. Med. 191, 771–780 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Madsen, L. et al. Mice lacking all conventional MHC class II genes. Proc. Natl Acad. Sci. USA 96, 10338–10343 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Meijlink, M. A. et al. 6′′-modifed α-GalCer-peptide conjugate vaccine candidates protect against liver-stage malaria. RSC Chem. Biol. 3, 551–560 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Benedict, M. Q. in The Molecular Biology of Insect Disease Vectors (eds Crampton, J. M. et al.) Ch. 1 (Chapman & Hall, 1997).

  59. Kimura, K. et al. CD8+ T cells specific for a malaria cytoplasmic antigen form clusters around infected hepatocytes and are protective at the liver stage of infection. Infect. Immun. 81, 3825–3834 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Cockburn, I. A. et al. Dendritic cells and hepatocytes use distinct pathways to process protective antigen from Plasmodium in vivo. PLoS Pathog. 7, e1001318 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Beattie, L. et al. A transcriptomic network identified in uninfected macrophages responding to inflammation controls intracellular pathogen survival. Cell Host Microbe 14, 357–368 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Chatzileontiadou, D. S. M., Szeto, C., Jayasinghe, D. & Gras, S. Protein purification and crystallization of HLA-A02:01 in complex with SARS-CoV-2 peptides. STAR Protoc. 2, 100635 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Farrand, K. et al. Using full-spectrum flow cytometry to phenotype memory T and NKT cell subsets with optimized tissue-specific preparation protocols. Curr. Protoc. 2, e482 (2022).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the BRF at the Peter Doherty Institute, and the BRU and the Hugh Green Cytometry Centre at the Malaghan Institute of Medical Research for technical support. We thank D. Bowen for support of K.E., D. Godfrey for the CD1d–PBS-44 tetramers and the NIH tetramer core facility for the CD1d–PBS-57 tetramers. This work was supported by funding from the New Zealand Ministry of Business Innovation and Employment (contract RTVU1603 to Victoria University of Wellington) and the New Zealand Health Research Council (contract HRC-20/569 to Victoria University of Wellington and HRC14/1003 Independent Research Organisation Fund to the Malaghan Institute). K.C.Y.P. is supported by the Monash Graduate Scholarship and the Monash University MNHS Faculty International Tuition Scholarship. D.F.R. was supported by the National Health and Medical Research Council of Australia (NHMRC) 1139486, S.G. by NHMRC 1159272, P.B. and K.E. by NHMRC 1146677, W.R.H. by NHMRC 1154457, and W.R.H. and L.E.H. by NHMRC 2012701.

Author information

Authors and Affiliations

Authors

Contributions

W.R.H., I.F.H, G.F.P, D.F.-R., L.E.H., D.F.A. and M.G. conceived the idea and designed the outline of the research. A.C., I.A.C., K.Y. and G.I.M. provided sporozoites for infection studies. K.C.Y.P., J.L.N. and J.R. carried out affinity measurements of 2C12 TCR–CD1d–αGCB by surface plasmon resonance. W.R.H., I.F.H., G.F.P., L.E.H. and M.G. wrote the manuscript. J.L.N., S.L.D., S.T.S.C, R.J.A., B.J.C. A.J.M. O.K.B. and J.J.M. wrote Methods sections. All authors contributed to reviewing and editing the manuscript and to relevant discussions. M.G., K.J.F., O.K.B., J.C.M., M.M., L.E.H., J.J.M., Y.C.C. and Z.G. vaccinated mice, analyzed T cell responses and examined protection. S.L.D., M.G., J.J.M., S.T.S.C, R.J.A., B.J.C. and A.M. prepared vaccines and vaccine components. S.G. produced Kb tetramers. C.X. produced CD1d–PBS-44 tetramers. K.E. and P.B. contributed to analysis of liver DC.

Corresponding authors

Correspondence to Gavin F. Painter, Ian F. Hermans or William R. Heath.

Ethics declarations

Competing interests

M.G., L.E.H., R.J.A., B.J.C., A.J.M. I.F.H., W.R.H. and G.F.P. are inventors on a patent application (WO2023121483A1) submitted by Victoria University of Wellington subsidiary Victoria Link Limited that covers the production of tissue-resident memory T cells with mRNA vaccines.

Peer review

Peer review information

Nature Immunology thanks Rafick Sekaly and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Jamie D. K. Wilson, in collaboration with the Nature Immunology team.

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 Vaccine-induced activation of splenic NKT cells.

Male B6 mice were transferred OT-I.CD45.1 cells one day before vaccination with mOVA vaccine alone, or mOVA with 80 pmol of indicated adjuvants. Data are combined from two independent experiments, giving n = 10 mice per group with the exception of the mOVA + αGCB group which contains 9 mice. a, Example flow cytometry plots showing expression of the TCR on splenic NKT cells at day 28. Gating shown in Supplementary data Fig. 4. b, Examples of CD69 (left) and PD-1 (right) expression on splenic NKT cells. c, Percentage TCRβ+ CD1d-tetramer+ spleen cells as gated in (a). Data are displayed as mean ± s.e.m. and individual mice (circles) and compared to the mOVA + αGC group by one-way ANOVA analysis with Tukey’s multiple comparison post-test. d, Geometric mean fluorescence intensity of PD-1 on NKT cells. Data are displayed as mean ± s.e.m. from two independent experiments and compared to the mOVA + αGC group by one-way ANOVA analysis with Tukey’s multiple comparison post-test. ****P < 0.0001.

Source data

Extended Data Fig. 2 Type I NKT 2C12 TCR binding affinity for mouse CD1d-αGCB.

Affinity measurement of soluble mouse type I NKT 2C12 TCR to mCD1d-αGCB using surface plasmon resonance (SPR). a, Equilibrium curves representative of one experiment performed in duplicate. b, Kd values are derived from duplicate runs from n = 3 independent experiments. Error bars on lower graph show mean ± SD.

Source data

Extended Data Fig. 3 Effect of vaccination on expression of costimulatory molecules on liver DC.

Female B6 mice were vaccinated with mOVA vaccine alone or mOVA with adjuvants αGC or αGCB. Livers were harvested 24 h later and DC (CD11c+ MHC II+ cells) assessed for relative expression of CD80 (a) and CD86 (b). Data are expressed as percent of maximum (% of max) determined by dividing the GMFI of each sample by the maximum mean GMFI of the highest mean of all groups from each experiment. Each circle depicts the values of an individual mouse (n = 10 biologically distinct samples combined from two independent experiments). Lines show mean ± s.e.m. Data (a, b) were compared by a one-way ANOVA with Tukey’s multiple comparison post-test (a, b). ****P < 0.0001.

Source data

Extended Data Fig. 4 The role of cytokines in responses to mRNA vaccination.

a, Male B6 mice were transferred OT-I.CD45.1 cells one day before vaccination with mOVA + αGCB. Groups of these mice were injected with mAb specific for blocking either IL-12p35, IL-15, GM-CSF or IFN-γR1, or with an isotype control mAb on days 0, 1 and 3 and then left to day 30 before OT-I memory T cell subsets were quantified in the liver. Gating parameters for OT-I cells are described in Supplementary data Fig. 1. Data are expressed as mean cell count ± s.e.m. for each subset. Individual values for each mouse are shown in Supplementary Fig. 8a. b, Female B6 mice or IL-15/ mice were transferred OT-I.CD45.1 cells one day before vaccination with mOVA + αGCB., and 36 days later OT-I memory T cell subsets were quantified in the liver. Mean cell count ± s.e.m. for each subset shown. Individual values for each mouse are shown in Supplementary Fig. 8b. c, Male B6 mice were OT-I.CD45.1 cells one day before vaccination mOVA + αGCB. Groups of these mice were injected with specific mAb for blocking IFNαR1 or an isotype control on days 0, 1 and 3 and then left to day 30 before OT-I memory T cell subsets were quantified in the liver. Mean cell count ± s.e.m. for each subset shown. Individual values for each mouse are shown in Supplementary data 8c. Data in a were log-transformed and compared to the isotype group by one-way ANOVA with Tukey’s multiple comparison post-test. Data in b and c were log-transformed and compared by two-sided unpaired Student’s t tests. Two independent experiments were performed for each panel (a,b, n = 10 mice per group; c, n = 7 mice per group). ****P < 0.0001. Bars indicating significance are coloured to correspond to each memory T cell subset.

Source data

Extended Data Fig. 5 Assessment of total anti-OVA IgG in sera 3 weeks following a prime-boost schedule.

Male B6 mice were vaccinated with mOVA vaccine alone (containing 5 µg mRNA), or mOVA adjuvanted with αGC or αGCB, on days 0 and 21, with sera collected 3 weeks after the boost analysed for total anti-OVA IgG by ELISA. Two independent experiments were combined to give 10 mice per group with the exception of the αGC group which contains 11 mice. Geometric mean ± 95 % confidence intervals for EC50 values are shown. Data were log-transformed and compared by one-way ANOVA with Tukey’s multiple comparison post-test. ****P < 0.0001.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–14 and Table 1.

Reporting Summary

Source data

Source Data Fig. 1

Statistical 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.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

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

Ganley, M., Holz, L.E., Minnell, J.J. et al. mRNA vaccine against malaria tailored for liver-resident memory T cells. Nat Immunol 24, 1487–1498 (2023). https://doi.org/10.1038/s41590-023-01562-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01562-6

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