A self-amplifying mRNA SARS-CoV-2 vaccine candidate induces safe and robust protective immunity in preclinical models

Giulietta Maruggi, Corey P Mallett, Jason W Westerbeck, Tiffany Chen, Giuseppe Lofano, Kristian Friedrich, Lin Qu, Jennifer Tong Sun, Josie McAuliffe, Amey Kanitkar, Kathryn T Arrildt, Kai-Fen Wang, Ian McBee, Deborah McCoy, Rebecca Terry, Alison Rowles, Maia Araujo Abrahim, Michael A Ringenberg, Malcolm J Gains, Catherine Spickler, Xuping Xie, Jing Zou, Pei-Yong Shi, Taru Dutt, Marcela Henao-Tamayo, Izabela Ragan, Richard A Bowen, Russell Johnson, Sandra Nuti, Kate Luisi, Jeffrey B Ulmer, Ann-Muriel Steff, Rashmi Jalah, Sylvie Bertholet, Alan H Stokes, Dong Yu, Giulietta Maruggi, Corey P Mallett, Jason W Westerbeck, Tiffany Chen, Giuseppe Lofano, Kristian Friedrich, Lin Qu, Jennifer Tong Sun, Josie McAuliffe, Amey Kanitkar, Kathryn T Arrildt, Kai-Fen Wang, Ian McBee, Deborah McCoy, Rebecca Terry, Alison Rowles, Maia Araujo Abrahim, Michael A Ringenberg, Malcolm J Gains, Catherine Spickler, Xuping Xie, Jing Zou, Pei-Yong Shi, Taru Dutt, Marcela Henao-Tamayo, Izabela Ragan, Richard A Bowen, Russell Johnson, Sandra Nuti, Kate Luisi, Jeffrey B Ulmer, Ann-Muriel Steff, Rashmi Jalah, Sylvie Bertholet, Alan H Stokes, Dong Yu

Abstract

RNA vaccines have demonstrated efficacy against SARS-CoV-2 in humans, and the technology is being leveraged for rapid emergency response. In this report, we assessed immunogenicity and, for the first time, toxicity, biodistribution, and protective efficacy in preclinical models of a two-dose self-amplifying messenger RNA (SAM) vaccine, encoding a prefusion-stabilized spike antigen of SARS-CoV-2 Wuhan-Hu-1 strain and delivered by lipid nanoparticles (LNPs). In mice, one immunization with the SAM vaccine elicited a robust spike-specific antibody response, which was further boosted by a second immunization, and effectively neutralized the matched SARS-CoV-2 Wuhan strain as well as B.1.1.7 (Alpha), B.1.351 (Beta) and B.1.617.2 (Delta) variants. High frequencies of spike-specific germinal center B, Th0/Th1 CD4, and CD8 T cell responses were observed in mice. Local tolerance, potential systemic toxicity, and biodistribution of the vaccine were characterized in rats. In hamsters, the vaccine candidate was well-tolerated, markedly reduced viral load in the upper and lower airways, and protected animals against disease in a dose-dependent manner, with no evidence of disease enhancement following SARS-CoV-2 challenge. Therefore, the SARS-CoV-2 SAM (LNP) vaccine candidate has a favorable safety profile, elicits robust protective immune responses against multiple SARS-CoV-2 variants, and has been advanced to phase 1 clinical evaluation (NCT04758962).

Keywords: SARS-CoV-2 vaccine; biodistribution; efficacy; immunogenicity; self-amplifying mRNA; spike antigen; toxicity.

Conflict of interest statement

Declaration of interests G.M., C.P.M., J.W., T.C., G.L., K.F., L.Q., J.T.S., J.M., A.K., K.A., K-F.W., I.M., R.T., A.R., M.A.R., A-M.S., R.Jo., S.N., R.J., K.L., S.B., J.B.U., A.H.S., and D.Y. are current or former employees of the GSK group of companies and may own GSK shares and/or restricted GSK shares. G.M., J.W., L.Q., K.L., J.B.U., and D.Y. are inventors on a patent application claiming subject matter related to the SARS-CoV-2 SAM vaccine candidates described herein. P.Y.S. is a member of the Scientific Advisory Boards of AbImmune and is Founder of FlaviTech. X.X. and P.-Y.S. have filed a patent on the reverse genetic system of SARS-CoV-2. M.A.A., M.G., and C.S. received compensation from GSK to perform the rat toxicity and biodistribution assays. The other authors declare no other competing interests.

Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Vaccine design and antigen characterization (A) Schematic representation of the SAM vector encoding the prefusion-stabilized SARS-CoV-2 spike (spikeFL-2P) protein (i.e., SARS-CoV-2 SAM). S1, S2, transmembrane (TM), and cytoplasmic (CT) domains, K986P, V987P, and GSAS mutations are indicated. (B) Percentage of spike-positive cells measured by intracellular flow cytometry 18 h after green fluorescent protein (GFP), prefusion-stabilized (spikeFL-2P), or wild-type (spikeFL) spike sequence SAM RNA electroporation of BHK cells, and intracellular staining with an anti-S2-specific antibody. (C) SAM-transfected BHK cells were also treated with (+) or without (−) BFA, and 18 h later, cell lysates were collected and analyzed by immunoblotting using anti-S2 or anti-actin antibodies. A recombinant spike protein was loaded as control. In addition, mouse C2C12 cells were transfected with SAM, treated with (+) or without (−) BFA, collected 18 h later, and surface-expressed spike protein was detected by flow cytometry using either the anti-S2-antibody (D) or incubated with soluble hACE2 and stained with an anti-hACE2 antibody (E). nsPs, non-structural proteins; UTR, untranslated regions; arrow, subgenomic promoter; kDa, kilodalton; S0, furin uncleaved full-length spike protein; S2, furin cleaved domain. Error bars represent SD of duplicate samples. Data shown are representative of at least two experiments.
Figure 2
Figure 2
Humoral responses elicited by SARS-CoV-2 SAM (LNP) vaccine in mice Pseudovirus-based spike 50% neutralization titers (pVNT50) of immunized mice sera or human COVID-19 convalescent sera (HCS) against Wuhan (A), Alpha (B.1.1.7) (C), Beta (B.1.351) (D), or Delta (B.1.617.2) (E) spike sequences. A panel of 22 HCS was tested against Wuhan, Alpha, and Beta, whereas a panel of 21 HCS (same panel minus one subject) was tested against Delta. Mouse sera were collected 3 weeks after the first vaccination (3wp1) and 2 weeks after the second vaccination (2wp2). (B) SARS-CoV-2 50% virus neutralization titers (VNT50) measured with a mNeonGreen reporter virus-based assay in sera collected 2wp2 from immunized mice or in a panel of COVID-19 HCS (n = 22). The dotted line indicates the limit of detection. Geometric means of each group ±95% confidence interval are shown. Each dot represents individual samples. Bars under p value asterisks indicate significantly different groups comparisons with ∗∗, p < 0.01; ∗∗∗, p < 0.001; ∗∗∗∗, p < 0.0001.
Figure 3
Figure 3
Cellular responses elicited by SARS-CoV-2 SAM (LNP) vaccine in mice Splenocytes of BALB/c mice (n = 5) immunized i.m. with SAM vaccine or saline control (mock) were harvested at 2wp2, stimulated ex vivo with a peptide mix specific to spikeFL-2P, and analyzed for CD8+ (A and C) and CD4+ (B and D) T cell responses by flow cytometry. The stacked bars (A and B) indicate distribution of the Tc0/Tc1/Tc2/Tc17 cytotoxic cells within the total CD8+ and the Th0/Th1/Th2/Th17 T helper cells within the CD4+ spike-specific T cells (mean ± SEM). Boolean combinations for spike-specific CD8+ and CD4+ T cell cytokines from splenocytes of individual immunized mice were background subtracted using Pestle software, and pies were generated using SPICE software (C and D). Each pie slice represents a proportion of the total spike-specific CD8+ or CD4+ T cell responses for a unique sub-population comprising CD107a, IFN-γ, IL-2, or TNF-α, as indicated in the legend. Bars under p value asterisks indicate significantly different group comparisons with ∗, p < 0.05; ∗∗, p < 0.01; ∗∗∗, p < 0.001; ∗∗∗∗, p < 0.0001.
Figure 4
Figure 4
Viral loads from oropharyngeal swabs and respiratory tract tissues in SARS-CoV-2 SAM (LNP) vaccinated hamsters after challenge with SARS-CoV-2 virus (A and B) Oropharyngeal swabs were taken from all hamsters on 1, 2, 3, and 7 dpi, and viral titers were determined by plaque assay. (C–E) Virus titers were determined in nasal turbinates (C), cranial right (D), and caudal right (E) lung lobes of four female and four male hamsters from each group at 3 dpi. Bars represent GMT+95% CI. Asterisks above bars indicate statistically significant difference in viral titers between mock and vaccine groups (∗∗∗∗, p 

Figure 5

Histopathology following SARS-CoV-2 challenge of…

Figure 5

Histopathology following SARS-CoV-2 challenge of SARS-CoV-2 SAM (LNP) and mock vaccinated hamsters (A)…

Figure 5
Histopathology following SARS-CoV-2 challenge of SARS-CoV-2 SAM (LNP) and mock vaccinated hamsters (A) Lung pathology grading, with respect to inflammation (A) and centriacinar/bronchioloalveolar epithelial hyperplasia (B) in male and female hamsters necropsied at 3, 7, and 21 dpi with SARS-CoV-2 virus. (C) Representative lung sections from a naive male hamster showing the background level of atelectasis that can occur during necropsy. (D) Representative lung sections from mock vaccinated, SAM low dose (0.03μg), and SAM high dose (3μg) vaccinated male animals infected with SARS-CoV-2 virus and necropsied at 3, 7, and 21 dpi (histopathological features in females are consistent with those in males, females not shown). Sections are stained with H&E, and whole slide images were obtained using P250 scanner (3D Histech). Representative low-power (bottom left, scale bar 5 mm), medium-power (top, scale bar 200 μm), and higher power (bottom right, scale bar 50 μm) photomicrographs are shown from each group. “#,” hyperplasia of type II pneumocytes and bronchiolization of alveoli; “G,” hyperplasia of goblet cells within bronchial epithelium; “H,” areas of alveolar wall necrosis and hemorrhage within regions of inflammation. Further description of the histopathology at each time point is provided in the main text.

Figure 6

Repeated-dose toxicology assessment in SARS-CoV-2…

Figure 6

Repeated-dose toxicology assessment in SARS-CoV-2 SAM (LNP) vaccinated rats (A) Body temperature. Male…

Figure 6
Repeated-dose toxicology assessment in SARS-CoV-2 SAM (LNP) vaccinated rats (A) Body temperature. Male and female rats were immunized on three occasions (day 1, 15, and 29) with 12 μg of SARS-CoV-2 SAM (LNP) or saline. Rectal temperatures were measured 1 h before (Pre) and 2, 4, 6, 12, 18, 24, and 48 h post immunization. Results are shown as mean with SD. Statistical analysis: ∗, p 
All figures (7)
Similar articles
Cited by
References
    1. Sempowski G.D., Saunders K.O., Acharya P., Wiehe K.J., Haynes B.F. Pandemic preparedness: developing vaccines and therapeutic antibodies for COVID-19. Cell. 2020;181:1458–1463. doi: 10.1016/j.cell.2020.05.041. - DOI - PMC - PubMed
    1. WHO Draft landscape and tracker of COVID-19 candidate vaccines. 2021. https://www.who.int/publications/m/item/draft-landscape-of-covid-19-cand...
    1. Polack F.P., Thomas S.J., Kitchin N., Absalon J., Gurtman A., Lockhart S., Perez J.L., Perez Marc G., Moreira E.D., Zerbini C., et al. Safety and efficacy of the BNT162b2 mRNA covid-19 vaccine. N. Engl. J. Med. 2020;383:2603–2615. doi: 10.1056/NEJMoa2034577. - DOI - PMC - PubMed
    1. FDA FDA approves first COVID-19 vaccine. 2021. https://www.fda.gov/news-events/press-announcements/fda-approves-first-c...
    1. Hekele A., Bertholet S., Archer J., Gibson D.G., Palladino G., Brito L.A., Otten G.R., Brazzoli M., Buccato S., Bonci A., et al. Rapidly produced SAM((R)) vaccine against H7N9 influenza is immunogenic in mice. Emerg. Microbes Infect. 2013;2:e52. doi: 10.1038/emi.2013.54. - DOI - PMC - PubMed
Show all 49 references
Publication types
MeSH terms
Supplementary concepts
Associated data
[x]
Cite
Copy Download .nbib .nbib
Format: AMA APA MLA NLM
Figure 5
Figure 5
Histopathology following SARS-CoV-2 challenge of SARS-CoV-2 SAM (LNP) and mock vaccinated hamsters (A) Lung pathology grading, with respect to inflammation (A) and centriacinar/bronchioloalveolar epithelial hyperplasia (B) in male and female hamsters necropsied at 3, 7, and 21 dpi with SARS-CoV-2 virus. (C) Representative lung sections from a naive male hamster showing the background level of atelectasis that can occur during necropsy. (D) Representative lung sections from mock vaccinated, SAM low dose (0.03μg), and SAM high dose (3μg) vaccinated male animals infected with SARS-CoV-2 virus and necropsied at 3, 7, and 21 dpi (histopathological features in females are consistent with those in males, females not shown). Sections are stained with H&E, and whole slide images were obtained using P250 scanner (3D Histech). Representative low-power (bottom left, scale bar 5 mm), medium-power (top, scale bar 200 μm), and higher power (bottom right, scale bar 50 μm) photomicrographs are shown from each group. “#,” hyperplasia of type II pneumocytes and bronchiolization of alveoli; “G,” hyperplasia of goblet cells within bronchial epithelium; “H,” areas of alveolar wall necrosis and hemorrhage within regions of inflammation. Further description of the histopathology at each time point is provided in the main text.
Figure 6
Figure 6
Repeated-dose toxicology assessment in SARS-CoV-2 SAM (LNP) vaccinated rats (A) Body temperature. Male and female rats were immunized on three occasions (day 1, 15, and 29) with 12 μg of SARS-CoV-2 SAM (LNP) or saline. Rectal temperatures were measured 1 h before (Pre) and 2, 4, 6, 12, 18, 24, and 48 h post immunization. Results are shown as mean with SD. Statistical analysis: ∗, p 
All figures (7)

References

    1. Sempowski G.D., Saunders K.O., Acharya P., Wiehe K.J., Haynes B.F. Pandemic preparedness: developing vaccines and therapeutic antibodies for COVID-19. Cell. 2020;181:1458–1463. doi: 10.1016/j.cell.2020.05.041.
    1. WHO Draft landscape and tracker of COVID-19 candidate vaccines. 2021.
    1. Polack F.P., Thomas S.J., Kitchin N., Absalon J., Gurtman A., Lockhart S., Perez J.L., Perez Marc G., Moreira E.D., Zerbini C., et al. Safety and efficacy of the BNT162b2 mRNA covid-19 vaccine. N. Engl. J. Med. 2020;383:2603–2615. doi: 10.1056/NEJMoa2034577.
    1. FDA FDA approves first COVID-19 vaccine. 2021.
    1. Hekele A., Bertholet S., Archer J., Gibson D.G., Palladino G., Brito L.A., Otten G.R., Brazzoli M., Buccato S., Bonci A., et al. Rapidly produced SAM((R)) vaccine against H7N9 influenza is immunogenic in mice. Emerg. Microbes Infect. 2013;2:e52. doi: 10.1038/emi.2013.54.
    1. Geall A.J., Verma A., Otten G.R., Shaw C.A., Hekele A., Banerjee K., Cu Y., Beard C.W., Brito L.A., Krucker T., et al. Nonviral delivery of self-amplifying RNA vaccines. Proc. Natl. Acad. Sci. U S A. 2012;109:14604–14609. doi: 10.1073/pnas.1209367109.
    1. Pepini T., Pulichino A.M., Carsillo T., Carlson A.L., Sari-Sarraf F., Ramsauer K., Debasitis J.C., Maruggi G., Otten G.R., Geall A.J., et al. Induction of an IFN-mediated antiviral response by a self-amplifying RNA vaccine: implications for vaccine design. J. Immunol. 2017;198:4012–4024. doi: 10.4049/jimmunol.1601877.
    1. Vogel A.B., Lambert L., Kinnear E., Busse D., Erbar S., Reuter K.C., Wicke L., Perkovic M., Beissert T., Haas H., et al. Self-amplifying RNA vaccines give equivalent protection against influenza to mRNA vaccines but at much lower doses. Mol. Ther. 2018;26:446–455. doi: 10.1016/j.ymthe.2017.11.017.
    1. Erasmus J.H., Khandhar A.P., Guderian J., Granger B., Archer J., Archer M., Gage E., Fuerte-Stone J., Larson E., Lin S., et al. A nanostructured lipid carrier for delivery of a replicating viral RNA provides single, low-dose protection against zika. Mol. Ther. 2018 doi: 10.1016/j.ymthe.2018.07.010.
    1. de Alwis R., Gan E.S., Chen S., Leong Y.S., Tan H.C., Zhang S.L., Yau C., Low J.G.H., Kalimuddin S., Matsuda D., et al. A single dose of self-transcribing and replicating RNA-based SARS-CoV-2 vaccine produces protective adaptive immunity in mice. Mol. Ther. 2021;29:1970–1983. doi: 10.1016/j.ymthe.2021.04.001.
    1. Sanders N.N. Low-dose single-shot COVID-19 mRNA vaccines lie ahead. Mol. Ther. 2021;29:1944–1945. doi: 10.1016/j.ymthe.2021.05.003.
    1. Wu F., Zhao S., Yu B., Chen Y.M., Wang W., Song Z.G., Hu Y., Tao Z.W., Tian J.H., Pei Y.Y., et al. A new coronavirus associated with human respiratory disease in China. Nature. 2020;579:265–269. doi: 10.1038/s41586-020-2008-3.
    1. Yi C., Sun X., Ye J., Ding L., Liu M., Yang Z., Lu X., Zhang Y., Ma L., Gu W., et al. Key residues of the receptor binding motif in the spike protein of SARS-CoV-2 that interact with ACE2 and neutralizing antibodies. Cell. Mol. Immunol. 2020;17:621–630. doi: 10.1038/s41423-020-0458-z.
    1. Yan R., Zhang Y., Li Y., Xia L., Guo Y., Zhou Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science. 2020;367:1444–1448. doi: 10.1126/science.abb2762.
    1. Jackson C.B., Farzan M., Chen B., Choe H. Mechanisms of SARS-CoV-2 entry into cells. Nat. Rev. Mol. Cell Biol. 2021 doi: 10.1038/s41580-021-00418-x.
    1. Wrapp D., Wang N., Corbett K.S., Goldsmith J.A., Hsieh C.L., Abiona O., Graham B.S., McLellan J.S. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. 2020;367:1260–1263. doi: 10.1126/science.abb2507.
    1. Tseng C.T., Sbrana E., Iwata-Yoshikawa N., Newman P.C., Garron T., Atmar R.L., Peters C.J., Couch R.B. Immunization with SARS coronavirus vaccines leads to pulmonary immunopathology on challenge with the SARS virus. PLoS One. 2012;7:e35421. doi: 10.1371/journal.pone.0035421.
    1. Bolles M., Deming D., Long K., Agnihothram S., Whitmore A., Ferris M., Funkhouser W., Gralinski L., Totura A., Heise M., Baric R.S. A double-inactivated severe acute respiratory syndrome coronavirus vaccine provides incomplete protection in mice and induces increased eosinophilic proinflammatory pulmonary response upon challenge. J. Virol. 2011;85:12201–12215. doi: 10.1128/JVI.06048-11.
    1. Muruato A.E., Fontes-Garfias C.R., Ren P., Garcia-Blanco M.A., Menachery V.D., Xie X., Shi P.Y. A high-throughput neutralizing antibody assay for COVID-19 diagnosis and vaccine evaluation. Nat. Commun. 2020;11:4059. doi: 10.1038/s41467-020-17892-0.
    1. Jhun H., Park H.Y., Hisham Y., Song C.S., Kim S. SARS-CoV-2 Delta (B.1.617.2) variant: a unique T478K mutation in receptor binding motif (RBM) of spike gene. Immune Netw. 2021;21:e32. doi: 10.4110/in.2021.21.e32.
    1. Sallusto F., Lanzavecchia A., Araki K., Ahmed R. From vaccines to memory and back. Immunity. 2010;33:451–463. doi: 10.1016/j.immuni.2010.10.008.
    1. Imai M., Iwatsuki-Horimoto K., Hatta M., Loeber S., Halfmann P.J., Nakajima N., Watanabe T., Ujie M., Takahashi K., Ito M., et al. Syrian hamsters as a small animal model for SARS-CoV-2 infection and countermeasure development. Proc. Natl. Acad. Sci. U S A. 2020;117:16587–16595. doi: 10.1073/pnas.2009799117.
    1. Chan J.F., Zhang A.J., Yuan S., Poon V.K., Chan C.C., Lee A.C., Chan W.M., Fan Z., Tsoi H.W., Wen L., et al. Simulation of the clinical and pathological manifestations of coronavirus disease 2019 (COVID-19) in a golden Syrian hamster model: implications for disease pathogenesis and transmissibility. Clin. Infect. Dis. 2020;71:2428–2446. doi: 10.1093/cid/ciaa325.
    1. EMA-CPMP Note for guidance on preclinical pharmacological and toxicological testing of vaccines (CPMP/SWP/465/95) 1997.
    1. WHO WHO guidelines on nonclinical evaluation of vaccines. 2003.
    1. Astakhova N.M., Perelygin A.A., Zharkikh A.A., Lear T.L., Coleman S.J., MacLeod J.N., Brinton M.A. Characterization of equine and other vertebrate TLR3, TLR7, and TLR8 genes. Immunogenetics. 2009;61:529–539. doi: 10.1007/s00251-009-0381-z.
    1. Baden L.R., El Sahly H.M., Essink B., Kotloff K., Frey S., Novak R., Diemert D., Spector S.A., Rouphael N., Creech C.B., et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N. Engl. J. Med. 2020 doi: 10.1056/NEJMoa2035389.
    1. Turner J.S., O'Halloran J.A., Kalaidina E., Kim W., Schmitz A.J., Zhou J.Q., Lei T., Thapa M., Chen R.E., Case J.B., et al. SARS-CoV-2 mRNA vaccines induce persistent human germinal centre responses. Nature. 2021;596:109–113. doi: 10.1038/s41586-021-03738-2.
    1. Wu K., Werner A.P., Moliva J.I., Koch M., Choi A., Stewart-Jones G.B.E., Bennett H., Boyoglu-Barnum S., Shi W., Graham B.S., et al. mRNA-1273 vaccine induces neutralizing antibodies against spike mutants from global SARS-CoV-2 variants. bioRxiv. 2021 doi: 10.1101/2021.01.25.427948.
    1. Channappanavar R., Fett C., Zhao J., Meyerholz D.K., Perlman S., Sandri-Goldin R.M. Virus-specific memory CD8 T cells provide substantial protection from lethal severe acute respiratory syndrome coronavirus infection. J. Virol. 2014;88:11034–11044. doi: 10.1128/JVI.01505-14.
    1. Grifoni A., Weiskopf D., Ramirez S.I., Mateus J., Dan J.M., Moderbacher C.R., Rawlings S.A., Sutherland A., Premkumar L., Jadi R.S., et al. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell. 2020;181:1489–1501 e1415. doi: 10.1016/j.cell.2020.05.015.
    1. Erasmus J.H., Khandhar A.P., O'Connor M.A., Walls A.C., Hemann E.A., Murapa P., Archer J., Leventhal S., Fuller J.T., Lewis T.B., et al. An Alphavirus-derived replicon RNA vaccine induces SARS-CoV-2 neutralizing antibody and T cell responses in mice and nonhuman primates. Sci. Transl. Med. 2020;12 doi: 10.1126/scitranslmed.abc9396.
    1. McKay P.F., Hu K., Blakney A.K., Samnuan K., Brown J.C., Penn R., Zhou J., Bouton C.R., Rogers P., Polra K., et al. Self-amplifying RNA SARS-CoV-2 lipid nanoparticle vaccine candidate induces high neutralizing antibody titers in mice. Nat. Commun. 2020;11:3523. doi: 10.1038/s41467-020-17409-9.
    1. Meyer M., Wang Y., Edwards D., Smith G.R., Rubenstein A.B., Ramanathan P., Mire C.E., Pietzsch C., Chen X., Ge Y., et al. Attenuated activation of pulmonary immune cells in mRNA-1273-vaccinated hamsters after SARS-CoV-2 infection. J. Clin. Invest. 2021;131 doi: 10.1172/JCI148036.
    1. Klein S.L., Flanagan K.L. Sex differences in immune responses. Nat. Rev. Immunol. 2016;16:626–638. doi: 10.1038/nri.2016.90.
    1. Fischinger S., Boudreau C.M., Butler A.L., Streeck H., Alter G. Sex differences in vaccine-induced humoral immunity. Semin. Immunopathol. 2019;41:239–249. doi: 10.1007/s00281-018-0726-5.
    1. Kariko K., Muramatsu H., Ludwig J., Weissman D. Generating the optimal mRNA for therapy: HPLC purification eliminates immune activation and improves translation of nucleoside-modified, protein-encoding mRNA. Nucleic Acids Res. 2011;39:e142. doi: 10.1093/nar/gkr695.
    1. Zhong Z., McCafferty S., Opsomer L., Wang H., Huysmans H., De Temmerman J., Lienenklaus S., Portela Catani J.P., Combes F., Sanders N.N. Corticosteroids and cellulose purification improve, respectively, the in vivo translation and vaccination efficacy of sa-mRNAs. Mol. Ther. 2021;29:1370–1381. doi: 10.1016/j.ymthe.2021.01.023.
    1. Zhong Z., Portela Catani J.P., Mc Cafferty S., Couck L., Van Den Broeck W., Gorle N., Vandenbroucke R.E., Devriendt B., Ulbert S., Cnops L., et al. Immunogenicity and protection efficacy of a naked self-replicating mRNA-based zika virus vaccine. Vaccines (Basel) 2019;7 doi: 10.3390/vaccines7030096.
    1. Stokes A., Pion J., Binazon O., Laffont B., Bigras M., Dubois G., Blouin K., Young J.K., Ringenberg M.A., Ben Abdeljelil N., et al. Nonclinical safety assessment of repeated administration and biodistribution of a novel rabies self-amplifying mRNA vaccine in rats. Regul. Toxicol. Pharmacol. 2020;113:104648. doi: 10.1016/j.yrtph.2020.104648.
    1. Segal L., Morelle D., Kaaber K., Destexhe E., Garcon N. Non-clinical safety assessment of single and repeated intramuscular administration of a human papillomavirus-16/18 vaccine in rabbits and rats. J. Appl. Toxicol. 2015;35:1577–1585. doi: 10.1002/jat.3131.
    1. Giordano G., Segal L., Prinsen M., Wijnands M.V., Garcon N., Destexhe E. Non-clinical safety assessment of single and repeated administration of gE/AS01 zoster vaccine in rabbits. J. Appl. Toxicol. 2017;37:132–141. doi: 10.1002/jat.3329.
    1. Huysmans H., Zhong Z., De Temmerman J., Mui B.L., Tam Y.K., Mc Cafferty S., Gitsels A., Vanrompay D., Sanders N.N. Expression kinetics and innate immune response after electroporation and LNP-mediated delivery of a self-amplifying mRNA in the skin. Mol. Ther. Nucleic Acids. 2019;17:867–878. doi: 10.1016/j.omtn.2019.08.001.
    1. Pollock K.M., Cheeseman H.M., Szubert A.J., Libri V., Boffito M., Owen D., Bern H., O’Hara J., McFarlane L.R., Lemm N., et al. Safety and immunogenicity of a self-amplifying RNA vaccine against COVID-19: COVAC1, a phase I, dose-ranging trial.
    1. Jeffs L.B., Palmer L.R., Ambegia E.G., Giesbrecht C., Ewanick S., MacLachlan I. A scalable, extrusion-free method for efficient liposomal encapsulation of plasmid DNA. Pharm. Res. 2005;22:362–372. doi: 10.1007/s11095-004-1873-z.
    1. Lou G., Anderluzzi G., Schmidt S.T., Woods S., Gallorini S., Brazzoli M., Giusti F., Ferlenghi I., Johnson R.N., Roberts C.W., et al. Delivery of self-amplifying mRNA vaccines by cationic lipid nanoparticles: the impact of cationic lipid selection. J. Control. Release. 2020;325:370–379. doi: 10.1016/j.jconrel.2020.06.027.
    1. Ragan I.K., Hartson L.M., Dutt T.S., Obregon-Henao A., Maison R.M., Gordy P., Fox A., Karger B.R., Cross S.T., Kapuscinski M.L., et al. A whole virion vaccine for COVID-19 produced via a novel inactivation method and preliminary demonstration of efficacy in an animal challenge model. Vaccines (Basel) 2021;9 doi: 10.3390/vaccines9040340.
    1. WHO WHO guidelines on nonclinical evaluation of vaccines. 2005.
    1. FGF Industry Considerations for plasmid DNA vaccines for infectious disease indications. 2007.

Source: PubMed

3
Abonneren