Proof-of-concept of a low-dose unmodified mRNA-based rabies vaccine formulated with lipid nanoparticles in human volunteers: A phase 1 trial

Cassandra Aldrich, Isabel Leroux-Roels, Katell Bidet Huang, Mihai Alexandru Bica, Edde Loeliger, Oliver Schoenborn-Kellenberger, Lisa Walz, Geert Leroux-Roels, Frank von Sonnenburg, Lidia Oostvogels, Cassandra Aldrich, Isabel Leroux-Roels, Katell Bidet Huang, Mihai Alexandru Bica, Edde Loeliger, Oliver Schoenborn-Kellenberger, Lisa Walz, Geert Leroux-Roels, Frank von Sonnenburg, Lidia Oostvogels

Abstract

Introduction: In a first-in-human study immune responses to rabies virus glycoprotein (RABV-G)-mRNA vaccine were dependent on the route of administration, necessitating specialized devices. Following successful preclinical studies with mRNA encapsulated in lipid nanoparticles (LNP), we tested an mRNA-LNP formulation (CV7202).

Methods: In this phase 1, multi-center, controlled study in Belgium and Germany we enrolled 55 healthy 18-40-year-olds to receive intramuscular injections of 5 μg (n = 10), 1 μg (n = 16), or 2 μg (n = 16) CV7202 on Day 1; subsets (n = 8) of 1 μg and 2 μg groups received second doses on Day 29. Controls (n = 10) received rabies vaccine, Rabipur, on Days 1, 8 and 29. Safety and reactogenicity were assessed up to 28 days post-vaccination using diary cards; immunogenicity was measured as RABV-G-specific neutralizing titers (VNT) by RFFIT and IgG by ELISA.

Results: As initially tested doses of 5 μg CV7202 elicited unacceptably high reactogenicity we subsequently tested 1 and 2 μg doses which were better tolerated. No vaccine-related serious adverse events or withdrawals occurred. Low, dose-dependent VNT responses were detectable from Day 15 and by Day 29%, 31% and 22% of 1, 2 and 5 μg groups, respectively, had VNTs ≥ 0·5 IU/mL, considered an adequate response by the WHO. After two 1 or 2 μg doses all recipients had titers ≥ 0.5 IU/mL by Day 43. Day 57 GMTs were not significantly lower than those with Rabipur, which elicited adequate responses in all vaccinees after two doses. CV7202-elicited VNT were significantly correlated with RABV-G-specific IgG antibodies (r2 = 0.8319, p < 0.0001).

Conclusions: Two 1 μg or 2 μg doses of CV7202 were well tolerated and elicited rabies neutralizing antibody responses that met WHO criteria in all recipients, but 5 μg had unacceptable reactogenicity for a prophylactic vaccine. ClinicalTrials.gov Identifier: NCT03713086.

Keywords: Lipid nanoparticles; Rabies; Vaccine; mRNA.

Conflict of interest statement

Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Copyright © 2020. Published by Elsevier Ltd.

Figures

Fig. 1
Fig. 1
Trial profile.
Fig. 2
Fig. 2
Geometric mean virus neutralizing titers (with 95% CI) in the four study groups after immunization (indicated by arrows) with CV7202 or Rabipur. Dashed line indicates level considered adequate by the WHO (0.5 IU/mL).
Fig. 3
Fig. 3
Responder rates (percentages of each group with a VNT ≥ 0.5 IU/mL) in the four study groups after immunization with CV7202 or Rabipur. Rates represent the numbers of participants achieving the protective VNT of 0.5 IU/mL. The 1 and 2 μg CV7202 groups consisted of 16 participants each for Days 8, 15 and 29, and 8 participants each for Days 36, 43 and 57. The 5 μg CV7202 group consisted of 10 participants for Days 8 and 15, 9 participants for Days 29, 36, 43 and 57. The Rabipur group had 10 participants at each timepoint.
Fig. 4
Fig. 4
GMTs (with 95% CI) of RABV-G-specific Ig responses assessed by ELISA. IgG concentrations after immunization with one (red arrow) or two (open arrow) doses of CV7202 or three doses of Rabipur (blue arrows). Dotted lines indicate LLOQ. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
Fig. 5
Fig. 5
Correlation of titers of RABV-G-specific neutralizing activity (VNT) and IgG antibodies after one or two doses of CV7202.

References

    1. Gouglas D, Christodoulou M, Plotkin SA, Hatchett R. CEPI: driving progress towards epidemic preparedness and response. Epidemiol Rev 2019; pii: mxz012.
    1. Chinazzi M., Davis J.T., Ajelli M., et al. The effect of travel restrictions on the spread of the 2019 novel coronavirus (COVID-19) outbreak. Science. 2020;368:395–400.
    1. Messina J.P., Brady O.J., Golding N., Kraemer M.U.G., Wint G.R.W., Ray S.E., et al. The current and future global distribution and population at risk of dengue. Nat Microbiol. 2019;4:1508–1515.
    1. Weaver S.C., Lecuit M. Chikungunya virus and the global spread of a mosquito-borne disease. N Engl J Med. 2015;372:1231–1239.
    1. Petersen E., Wilson M.E., Touch S., McCloskey B., Mwaba P., Bates M., et al. Rapid spread of Zika virus in the Americas—implications for public health preparedness for mass gatherings at the 2016 Brazil Olympic Games. Int J Infect Dis. 2016;44:11–15.
    1. Jackson N.A.C., Kester K.E., Casimiro D., Gurunathan S., DeRosa F. The promise of mRNA vaccines: a biotech and industrial perspective. npj Vaccines. 2020;5:11.
    1. Rauch S., Lutz J., Kowalczyk A., Schlake T., Heidenreich R. RNActive® technology: generation and testing of stable and immunogenic mRNA vaccines. Methods Mol Biol. 2017;1499:89–107.
    1. Alberer M., Gnad-Vogt U., Hong H.S., Mehr K.T., Backert L., Finak G., et al. Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an open-label, non-randomised, prospective, first-in-human phase 1 clinical trial. Lancet. 2017;390:1511–1520.
    1. Lutz J., Lazzaro S., Habbeddine M., Schmidt K.E., Baumhof P., Mui B.L., et al. Unmodified mRNA in LNPs constitutes a competitive technology for prophylactic vaccines. npj Vaccines. 2017;2:29.
    1. CDC. Toxicity grading scale for healthy adult and adolescent volunteers enrolled in preventive vaccine clinical trials (September 2007) Available at: Accessed 28 February 2020.
    1. WHO. Rabies vaccines WHO position paper. Wkly Epidemiol Rec 2007; 82:425–36.
    1. Hopkins R.J., Daczkowski N.F., Kaptur P.E., et al. Randomized, double-blind, placebo-controlled, safety and immunogenicity study of 4 formulations of Anthrax Vaccine Adsorbed plus CPG 7909 (AV7909) in healthy adult volunteers. Vaccine. 2013;31:3051–3058.
    1. Kløverpris H.N., Akil Jackson A., Handley A., et al. Non-immunogenicity of overlapping Gag peptides pulsed on autologous cells after vaccination of HIV infected individuals. PLoS ONE. 2013;8
    1. Sieg A., Arab L., Schlierf G., Stiehl A., Kommerell B. Die Prävalenz des Gilbert-Syndroms in Deutschland. Prevalence of Gilbert's syndrome in GermanyDtsch Med Wochenschr. 1987;112:1206–1208.
    1. Strassburg C.P. Hyperbilirubinemia syndromes (Gilbert-Meulengracht, Crigler-Najjar, Dubin-Johnson, and Rotor syndrome) Best Pract Res Clin Gastroenterol. 2010;24:555–571.
    1. Wagner K.H., Shiels R.G., Lang C.A., Khoei N.S., Bulmer A.C. Diagnostic criteria and contributors to Gilbert’s syndrome. Crit Rev Clin Lab Sci. 2018;55:129–139.
    1. Mulligan M.J., Lyke K.E., Kitchin N., et al. Phase I/II study of COVID-19 RNA vaccine BNT162b1 in adults. Nature. 2020;586:589–593.
    1. Jackson L.A., Anderson E.J., Rouphael N.G., et al. An mRNA vaccine against SARS-CoV-2 - preliminary report. New Engl J Med. 2020;383:1920–1931.
    1. McNamara M.A., Nair S.K., Holl E.K. RNA-based vaccines in cancer immunotherapy. J Immunol Res. 2015;2015
    1. Sebastian M., Schröder A., Scheel B., et al. A phase I/IIa study of the mRNA-based cancer immunotherapy CV9201 in patients with stage IIIB/IV non-small cell lung cancer. Cancer Immunol Immunother. 2019;68:799–812.
    1. Wang F., Kream R.M., Stefano G.B. An evidence based perspective on mRNA-SARS-CoV-2 vaccine development. Med Sci Monit. 2020;26
    1. Pardi N., Hogan M.J., Porter F.W., Weissman D. mRNA vaccines - a new era in vaccinology. Nat Rev Drug Discov. 2018;17:261–279.
    1. Reichmuth A.M., Oberli M.A., Jaklenec A., Langer R., Blankschtein D. mRNA vaccine delivery using lipid nanoparticles. Ther Deliv. 2016;7:319–334.
    1. Armbruster N., Jasny E., Petsch B. Advances in RNA vaccines for preventive indications: A case study of a vaccine against rabies. Vaccines (Basel) 2019;7:pii: E132.

Source: PubMed

3
Abonner