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:

A malaria vaccine adjuvant based on recombinant antigen binding to liposomes

Subjects

Abstract

Pfs25 is a malaria transmission-blocking vaccine antigen candidate, but its apparently limited immunogenicity in humans has hindered clinical development. Here, we show that recombinant, polyhistidine-tagged (his-tagged) Pfs25 can be mixed at the time of immunization with pre-formed liposomes containing cobalt porphyrin–phospholipid, resulting in spontaneous nanoliposome antigen particleization (SNAP). Antigens are stably presented in uniformly orientated display via his-tag insertion in the cobalt porphyrin–phospholipid bilayer, without covalent modification or disruption of antigen conformation. SNAP immunization of mice and rabbits is well tolerated with minimal local reactogenicity, and results in orders-of-magnitude higher functional antibody generation compared with other ‘mix-and-inject’ adjuvants. Serum-stable antigen binding during transit to draining lymph nodes leads to enhanced antigen uptake by phagocytic antigen-presenting cells, with subsequent generation of long-lived, antigen-specific plasma cells. Seamless multiplexing with four additional his-tagged Plasmodium falciparum polypeptides induces strong and balanced antibody production, illustrating the simplicity of developing multistage particulate vaccines with SNAP immunization.

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: SNAP with his-tagged Pfs25.
Fig. 2: Pfs25 SNAP immunization in mice and rabbits.
Fig. 3: Mechanistic insights into SNAP immunization.
Fig. 4: Durability of the anti-Pfs25 IgG response with SNAP immunization.
Fig. 5: Immunization potency and local reactogenicity of SNAP compared with other adjuvants (all mixed before injection).
Fig. 6: Multiplexed SNAP with P. falciparum antigens.

Similar content being viewed by others

Data availability

All raw data are available upon request.

References

  1. WHO World Malaria Report 2017 (World Health Organization, 2017).

  2. Nunes, J. K. et al. Development of a transmission-blocking malaria vaccine: progress, challenges, and the path forward. Vaccine 32, 5531–5539 (2014).

    Article  Google Scholar 

  3. Birkett, A. J. Status of vaccine research and development of vaccines for malaria. Vaccine 34, 2915–2920 (2016).

    Article  Google Scholar 

  4. Barr, P. J. et al. Recombinant Pfs25 protein of Plasmodium falciparum elicits malaria transmission-blocking immunity in experimental animals. J. Exp. Med. 174, 1203–1208 (1991).

    Article  CAS  Google Scholar 

  5. Kaslow, D. C. et al. A vaccine candidate from the sexual stage of human malaria that contains EGF-like domains. Nature 333, 74–76 (1988).

    Article  CAS  Google Scholar 

  6. Kaslow, D. C. et al. Saccharomyces cerevisiae recombinant Pfs25 adsorbed to alum elicits antibodies that block transmission of Plasmodium falciparum. Infect. Immun. 62, 5576–5580 (1994).

    CAS  Google Scholar 

  7. Kumar, R., Angov, E. & Kumar, N. Potent malaria transmission-blocking antibody responses elicited by Plasmodium falciparum Pfs25 expressed in Escherichia coli after successful protein refolding. Infect. Immun. 82, 1453–1459 (2014).

    Article  Google Scholar 

  8. Gregory, J. A. et al. Algae-produced Pfs25 elicits antibodies that inhibit malaria transmission. PLoS One 7, e37179 (2012).

    Article  CAS  Google Scholar 

  9. Mlambo, G., Kumar, N. & Yoshida, S. Functional immunogenicity of baculovirus expressing Pfs25, a human malaria transmission-blocking vaccine candidate antigen. Vaccine 28, 7025–7029 (2010).

    Article  CAS  Google Scholar 

  10. Lee, S.-M. et al. Assessment of Pfs25 expressed from multiple soluble expression platforms for use as transmission-blocking vaccine candidates. Malaria J. 15, 405 (2016).

    Article  Google Scholar 

  11. Wu, Y. et al. Phase 1 trial of malaria transmission blocking vaccine candidates Pfs25 and Pvs25 formulated with Montanide ISA 51. PLoS One 3, e2636 (2008).

    Article  Google Scholar 

  12. Malkin, E. M. et al. Phase 1 vaccine trial of Pvs25H: a transmission blocking vaccine for Plasmodium vivax malaria. Vaccine 23, 3131–3138 (2005).

    Article  CAS  Google Scholar 

  13. Radtke, A. J. et al. Adjuvant and carrier protein-dependent T-cell priming promotes a robust antibody response against the Plasmodium falciparum Pfs25 vaccine candidate. Sci. Rep. 7, 40312 (2017).

    Article  CAS  Google Scholar 

  14. Shimp, R. L. Jr et al. Development of a Pfs25-EPA malaria transmission blocking vaccine as a chemically conjugated nanoparticle. Vaccine 31, 2954–2962 (2013).

    Article  CAS  Google Scholar 

  15. Miyata, T. et al. Plasmodium vivax ookinete surface protein Pvs25 linked to cholera toxin B subunit induces potent transmission-blocking immunity by intranasal as well as subcutaneous immunization. Infect. Immun. 78, 3773–3782 (2010).

    Article  CAS  Google Scholar 

  16. Gregory, J. A., Topol, A. B., Doerner, D. Z. & Mayfield, S. Alga-produced cholera toxin-Pfs25 fusion proteins as oral vaccines. Appl. Environ. Microbiol. 79, 3917–3925 (2013).

    Article  CAS  Google Scholar 

  17. Kumar, R. et al. Nanovaccines for malaria using Plasmodium falciparum antigen Pfs25 attached gold nanoparticles. Vaccine 33, 5064–5071 (2015).

    Article  CAS  Google Scholar 

  18. Kumar, R. et al. Potent functional immunogenicity of Plasmodium falciparum transmission-blocking antigen (Pfs25) delivered with nanoemulsion and porous polymeric nanoparticles. Pharm. Res. 32, 3827–3836 (2015).

    Article  CAS  Google Scholar 

  19. Jones, R. M. et al. A plant-produced Pfs25 VLP malaria vaccine candidate induces persistent transmission blocking antibodies against Plasmodium falciparum in immunized mice. PLoS One 8, e79538 (2013).

    Article  Google Scholar 

  20. Goodman, A. L. et al. A viral vectored prime-boost immunization regime targeting the malaria Pfs25 antigen induces transmission-blocking activity. PLoS One 6, e29428 (2011).

    Article  CAS  Google Scholar 

  21. Li, Y. et al. Enhancing immunogenicity and transmission-blocking activity of malaria vaccines by fusing Pfs25 to IMX313 multimerization technology. Sci. Rep. 6, 18848 (2016).

    Article  CAS  Google Scholar 

  22. Brune, K. D. et al. Plug-and-display: decoration of virus-like particles via isopeptide bonds for modular immunization. Sci. Rep. 6, 19234 (2016).

    Article  CAS  Google Scholar 

  23. Shao, S. et al. Functionalization of cobalt porphyrin–phospholipid bilayers with his-tagged ligands and antigens. Nat. Chem. 7, 438–446 (2015).

    Article  CAS  Google Scholar 

  24. Lee, S.-M., Plieskatt, J. & King, C. R. Disulfide bond mapping of Pfs25, a recombinant malaria transmission blocking vaccine candidate. Anal. Biochem. 542, 20–23 (2018).

    Article  CAS  Google Scholar 

  25. Rüger, R., Müller, D., Fahr, A. & Kontermann, R. E. In vitro characterization of binding and stability of single-chain Fv Ni-NTA-liposomes. J. Drug Target. 14, 576–582 (2006).

    Article  Google Scholar 

  26. Platt, V. et al. Influence of multivalent nitrilotriacetic acid lipid−ligand affinity on the circulation half-life in mice of a liposome-attached His6-protein. Bioconj. Chem. 21, 892–902 (2010).

    Article  CAS  Google Scholar 

  27. Bale, S. et al. Covalent linkage of HIV-1 trimers to synthetic liposomes elicits improved B cell and antibody responses. J. Virol. 91, e00443-17 (2017).

    Article  Google Scholar 

  28. Scally, S. W. et al. Molecular definition of multiple sites of antibody inhibition of malaria transmission-blocking vaccine antigen Pfs25. Nat. Commun. 8, 1568 (2017).

    Article  Google Scholar 

  29. Beck, Z. et al. Differential immune responses to HIV-1 envelope protein induced by liposomal adjuvant formulations containing monophosphoryl lipid A with or without QS21. Vaccine 33, 5578–5587 (2015).

    Article  CAS  Google Scholar 

  30. Sauer, S. W. & Keim, M. E. Hydroxocobalamin: improved public health readiness for cyanide disasters. Ann. Emerg. Med. 37, 635–641 (2001).

    Article  CAS  Google Scholar 

  31. Kuzminski, A. M., Del Giacco, E. J., Allen, R. H., Stabler, S. P. & Lindenbaum, J. Effective treatment of cobalamin deficiency with oral cobalamin. Blood 92, 1191–1198 (1998).

    CAS  Google Scholar 

  32. Calabro, S. et al. Vaccine adjuvants Alum and MF59 induce rapid recruitment of neutrophils and monocytes that participate in antigen transport to draining lymph nodes. Vaccine 29, 1812–1823 (2011).

    Article  CAS  Google Scholar 

  33. Liang, F. et al. Vaccine priming is restricted to draining lymph nodes and controlled by adjuvant-mediated antigen uptake. Sci. Transl. Med. 9, eaal2094 (2017).

    Article  Google Scholar 

  34. Manh, T. P., Alexandre, Y., Baranek, T., Crozat, K. & Dalod, M. Plasmacytoid, conventional, and monocyte-derived dendritic cells undergo a profound and convergent genetic reprogramming during their maturation. Eur. J. Immunol. 43, 1706–1715 (2013).

    Article  CAS  Google Scholar 

  35. Ginhoux, F. et al. The origin and development of nonlymphoid tissue CD103+ DCs. J. Exp. Med. 206, 3115–3130 (2009).

    Article  CAS  Google Scholar 

  36. Edelson, B. T. et al. Peripheral CD103+ dendritic cells form a unified subset developmentally related to CD8α+ conventional dendritic cells. J. Exp. Med. 207, 823–836 (2010).

    Article  CAS  Google Scholar 

  37. Rooijen, N. V. & Sanders, A. Liposome mediated depletion of macrophages: mechanism of action, preparation of liposomes and applications. J. Immunol. Methods 174, 83–93 (1994).

    Article  Google Scholar 

  38. Allen, T. M., Austin, G. A., Chonn, A., Lin, L. & Lee, K. C. Uptake of liposomes by cultured mouse bone marrow macrophages: influence of liposome composition and size. Biochim. Biophys. Acta 1061, 56–64 (1991).

    Article  CAS  Google Scholar 

  39. Fan, Y. C., Sahdev, P., Ochyl, L. J., Akerberg, J. J. & Moon, J. J. Cationic liposome–hyaluronic acid hybrid nanoparticles for intranasal vaccination with subunit antigens. J. Control. Release 208, 121–129 (2015).

    Article  CAS  Google Scholar 

  40. Cunningham, A. L. et al. Efficacy of the herpes zoster subunit vaccine in adults 70 years of age or older. N. Engl. J. Med. 375, 1019–1032 (2016).

    Article  CAS  Google Scholar 

  41. Viswanathan, S., Rani, C., Vijay Anand, A. & Ho, J. A. Disposable electrochemical immunosensor for carcinoembryonic antigen using ferrocene liposomes and MWCNT screen-printed electrode. Biosens. Bioelectron. 24, 1984–1989 (2009).

    Article  CAS  Google Scholar 

  42. Doolan, D. L. & Hoffman, S. L. DNA-based vaccines against malaria: status and promise of the Multi-Stage Malaria DNA Vaccine Operation. Int. J. Parasitol. 31, 753–762 (2001).

    Article  CAS  Google Scholar 

  43. Peterson, M. G. et al. Integral membrane protein located in the apical complex of Plasmodium falciparum. Mol. Cell. Biol. 9, 3151–3154 (1989).

    Article  CAS  Google Scholar 

  44. Lobo, C. A., Konings, R. N. & Kumar, N. Expression of early gametocyte-stage antigens Pfg27 and Pfs16 in synchronized gametocytes and non-gametocyte producing clones of Plasmodium falciparum. Mol. Biochem. Parasitol. 68, 151–154 (1994).

    Article  CAS  Google Scholar 

  45. Farrance, C. E. et al. Antibodies to plant-produced Plasmodium falciparum sexual stage protein Pfs25 exhibit transmission blocking activity. Hum. Vaccin. 7, 191–198 (2011).

    Article  CAS  Google Scholar 

  46. Lee, S. M. et al. An N-terminal Pfs230 domain produced in baculovirus as a biological active transmission-blocking vaccine candidate. Clin. Vaccine Immunol. 24, e00140-17 (2017).

    Article  Google Scholar 

  47. Dutta, S. et al. High antibody titer against apical membrane antigen-1 is required to protect against malaria in the Aotus model. PLoS One 4, e8138 (2009).

    Article  Google Scholar 

  48. Genito, C. J. et al. Liposomes containing monophosphoryl lipid A and QS-21 serve as an effective adjuvant for soluble circumsporozoite protein malaria vaccine FMP013. Vaccine 35, 3865–3874 (2017).

    Article  CAS  Google Scholar 

  49. Miura, K. et al. Development and characterization of a standardized ELISA including a reference serum on each plate to detect antibodies induced by experimental malaria vaccines. Vaccine 26, 193–200 (2008).

    Article  CAS  Google Scholar 

  50. Quakyi, I. A. et al. The 230-kDa gamete surface protein of Plasmodium falciparum is also a target for transmission-blocking antibodies. J. Immunol. 139, 4213–4217 (1987).

    CAS  Google Scholar 

  51. Cheru, L. et al. The IC50 of anti-Pfs25 antibody in membrane-feeding assay varies among species. Vaccine 28, 4423–4429 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This study was supported by PATH’s Malaria Vaccine Initiative, and grants from the National Institutes of Health (R21AI122964 and DP5OD017898) and the intramural programme of the National Institute of Allergy and Infectious Diseases/NIH. The authors acknowledge assistance from G. Mlambo and A. Tripathi with immunofluorescence assays, and input from C. Alving and A. Birkett.

Author information

Authors and Affiliations

Authors

Contributions

W.-C.H., C.R.K., S.-M.L. and J.F.L conceived the project. W.-C.H., K.M., S.-M.L. and J.F.L. designed most of the experiments. W.-C.H. and J.F.L. wrote the manuscript. W.-C.H., C.L. and J.G. performed the animal experiments. W.-C.H., C.L., B.D., C.A.L. and K.M. performed and interpreted the ELISA and SMFA experiments. A.R. and J.O. performed transmission electron cryomicroscopy. W.-C.H., K.A.C., C.L., X.H. and B.S. produced and characterized the liposomes. X.H. performed the splenocyte studies. U.C. and W.-C.H. produced fluorescently labelled antigens. J.F. performed the circular dichroism studies. S.D. and S.-M.L. produced the antigens.

Corresponding author

Correspondence to Jonathan F. Lovell.

Ethics declarations

Competing interests

W.-C.H., C.R.K., S.-M.L., J.G. and J.F.L are named inventors on a patent application describing this technology. All other authors declare no competing financial interests.

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 Methods, Supplementary Figures 1–20 and Supplementary Tables 1–5

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huang, WC., Deng, B., Lin, C. et al. A malaria vaccine adjuvant based on recombinant antigen binding to liposomes. Nature Nanotech 13, 1174–1181 (2018). https://doi.org/10.1038/s41565-018-0271-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41565-018-0271-3

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