Lipid nanoparticle-encapsulated mRNA antibody provides long-term protection against SARS-CoV-2 in mice and hamsters

Yong-Qiang Deng, Na-Na Zhang, Yi-Fei Zhang, Xia Zhong, Sue Xu, Hong-Ying Qiu, Tie-Cheng Wang, Hui Zhao, Chao Zhou, Shu-Long Zu, Qi Chen, Tian-Shu Cao, Qing Ye, Hang Chi, Xiang-Hui Duan, Dan-Dan Lin, Xiao-Jing Zhang, Liang-Zhi Xie, Yu-Wei Gao, Bo Ying, Cheng-Feng Qin, Yong-Qiang Deng, Na-Na Zhang, Yi-Fei Zhang, Xia Zhong, Sue Xu, Hong-Ying Qiu, Tie-Cheng Wang, Hui Zhao, Chao Zhou, Shu-Long Zu, Qi Chen, Tian-Shu Cao, Qing Ye, Hang Chi, Xiang-Hui Duan, Dan-Dan Lin, Xiao-Jing Zhang, Liang-Zhi Xie, Yu-Wei Gao, Bo Ying, Cheng-Feng Qin

Abstract

Monoclonal antibodies represent important weapons in our arsenal to against the COVID-19 pandemic. However, this potential is severely limited by the time-consuming process of developing effective antibodies and the relative high cost of manufacturing. Herein, we present a rapid and cost-effective lipid nanoparticle (LNP) encapsulated-mRNA platform for in vivo delivery of SARS-CoV-2 neutralization antibodies. Two mRNAs encoding the light and heavy chains of a potent SARS-CoV-2 neutralizing antibody HB27, which is currently being evaluated in clinical trials, were encapsulated into clinical grade LNP formulations (named as mRNA-HB27-LNP). In vivo characterization demonstrated that intravenous administration of mRNA-HB27-LNP in mice resulted in a longer circulating half-life compared with the original HB27 antibody in protein format. More importantly, a single prophylactic administration of mRNA-HB27-LNP provided protection against SARS-CoV-2 challenge in mice at 1, 7 and even 63 days post administration. In a close contact transmission model, prophylactic administration of mRNA-HB27-LNP prevented SARS-CoV-2 infection between hamsters in a dose-dependent manner. Overall, our results demonstrate a superior long-term protection against SARS-CoV-2 conferred by a single administration of this unique mRNA antibody, highlighting the potential of this universal platform for antibody-based disease prevention and therapy against COVID-19 as well as a variety of other infectious diseases.

Trial registration: ClinicalTrials.gov NCT03829384.

Conflict of interest statement

C.-F.Q. and B.Y. have filed a patent related to the technology reported in this article. B.Y., X.Z., S.X., X.-H.D., D.-D.L., and X.-J.Z. are employee of Suzhou Abogen Biosciences. L.-Z.X has an ownership in Sinocelltech. The other authors have no conflicts of interest to declare.

© 2022. The Author(s).

Figures

Fig. 1. Rational design and characterization of…
Fig. 1. Rational design and characterization of the mRNA antibody.
a Design and encapsulation of an mRNA antibody mRNA-HB27-LNP. b HB27 antibody expression in Vero, 293 T and Expi293F cells at 24 h after transfection with mRNA-HB27 using ELISA assay. Data are shown as mean ± SEM. Data are analyzed by One-way ANOVA with multiple comparisons (****P < 0.0001). c Inhibition of SARS-CoV-2 infection in Vero cells at 24 h after transfection with 1 μg of mRNA-HB27 by Immunofluorescence assay. SARS-CoV-2 S protein was stained in green, and DAPI in blue. Scale bars, 100 μm. d, e The antibody concentration and NT50 titer of serum samples in mice. Briefly, female BALB/c mice (n = 4/group) were i.v. administrated with the indicated dose of mRNA-HB27-LNP or Placebo. Then, mice sera at 24 h post administration were measured by ELISA (d) and SARS-CoV-2 pseudovirus neutralization assay (e), respectively. Dotted lines indicate the limits of detection. Data are shown as mean ± SEM. f Intravenous injection of reporter mRNA-LNPs for in vivo imaging in mice. IVIS Spectrum image (6 h post-injection) of female BALB/c mice were injected with 10 μg of FLuc-encoding reporter mRNA-LNP by the intravenous route.
Fig. 2. Prophylactic efficacy of mRNA-HB27-LNP against…
Fig. 2. Prophylactic efficacy of mRNA-HB27-LNP against SARS-CoV-2 in mice.
a Experimental design. Briefly, groups of 8-month-old female BALB/c mice (n = 32) were i.v. administrated with a single dose of 1 mg/kg (purple), 0.2 mg/kg (blue), or 0.4 mg/kg (orange) of mRNA-HB27-LNP and Placebo (red) at 1 days before challenge with 6 × 103 PFU of MASCp36, and the clinical symptoms and mortality were recorded for 14 days. Mice (n = 3 per group) were sacrificed at 3 dpi for viral detection and histopathological analysis. b Survival curves of BALB/c mice administrated with the indicated dose of mRNA-HB27-LNP (n = 5 per group) and Placebo (n = 5). Data were analyzed by Wilcoxon log-rank survival test. (***P < 0.001, ****P < 0.0001). c viral sgRNA loads of lung tissues at 3 dpi were determined by RT-qPCR, respectively. Data are represented as mean ± SEM. Dashed lines represents limit of detection (n = 3 per group). Significance was calculated using Ordinary one-way ANOVA Multiple comparisons (****P < 0.0001). d Histopathological analysis of lung tissues at 3 dpi. Scale bar, 100 µm. e Experimental design. Briefly, the 8-month-old male BALB/c mice received i.v. administration of 1 mg/kg of mRNA-HB27-LNP (n = 5) and Placebo (n = 5), respectively. At 1 day post administration, mice were i.n. challenged with 1 × 104 PFU of SARS-CoV-2 Beta variant strain, and the lung tissues were collected at 5 days after challenge for detection of viral burden determination (by RT-qPCR) and histopathological analysis. f Viral sgRNA loads of lung tissues at 5 dpi were determined by RT-qPCR. Data are represented as mean ± SEM. Significance was calculated using unpaired t-test (****P < 0.0001). g Histopathological analysis of lung tissues at 5 dpi. Scale bar, 100 µm.
Fig. 3. mRNA-HB27-LNP prevents SARS-CoV-2 transmission in…
Fig. 3. mRNA-HB27-LNP prevents SARS-CoV-2 transmission in hamsters.
a Experimental design. Briefly, groups of 6–8-week-old female Syrian golden hamsters were i.v. injected with 0.3 or 1 mg/kg of mRNA-HB27-LNP (n = 2 per group) or placebo (n = 4). 24 h later, each of treatment hamster were transferred to a new cage and were cohoused with one index hamster in a one-to-one manner. The index hamsters (n = 8) were infected with 1 × 104 PFU of SARS-CoV-2 through the intranasal route at 1 h prior to cohouse. The lung tissues of hamster were collected at 4 dpi for following-up viral burden determination (by RT-qPCR) and histopathological analysis. b Viral sgRNA loads in lung tissues of hamsters at 4 dpi were determined by RT-qPCR. The dotted line in panel shows the limit of detection of the assay. c Histopathological analysis of each group of lung tissues at 5 dpi. Representative images from 16 hamsters are shown. Scale bar, 200 µm.
Fig. 4. mRNA-HB27-LNP provides a long-term protection…
Fig. 4. mRNA-HB27-LNP provides a long-term protection against SARS-CoV-2 challenge in mice.
a The antibody concentration of serum in mice by ELISA. Briefly, groups of 6–8-week-old ICR mice were i.v. administrated with a single dose of 1 mg/kg of HB27 (n = 4) and HB27-mRNA-LNP (n = 4), respectively. At indicated times post administration, sera of mice were measured by ELISA. Dotted lines indicate the limits of detection. b Analysis of antibody pharmacokinetics in serum after the i.v. administration with a single dose of HB27 and mRNA-HB27-LNP. Calculations were performed using WinNolin. c NT50 of serum in mice by VSV-based SARS-CoV-2 pseudovirus. Data are shown as mean ± SEM. Dashed lines represents limit of detection. d, e Experimental design. Briefly, groups of 8-month-old female BALB/c mice were i.v. administrated with a single dose of 1 mg /kg of HB27 or mRNA-HB27-LNP (n = 4 or 5) and Placebo (n = 5). Then at 7 days or 63 days post administration, mice were challenged with 6 × 103 PFU of MASCp36, respectively, and the clinical symptoms and mortality were recorded for 14 days. Survival curves of mice after lethal challenge by MASCp36 at 7 days (d) and 63 days (e) after the i.v. administration. Data were analyzed by Wilcoxon log-rank survival test (**P < 0.01).

References

    1. Hu B, Guo H, Zhou P, Shi ZL. Characteristics of SARS-CoV-2 and COVID-19. Nat. Rev. Microbiol. 2021;19:141–154. doi: 10.1038/s41579-020-00459-7.
    1. Dai L, Gao GF. Viral targets for vaccines against COVID-19. Nat. Rev. Immunol. 2021;21:73–82. doi: 10.1038/s41577-020-00480-0.
    1. Kumari, P., Rawat, K. & Saha, L. Pipeline pharmacological therapies in clinical trial for COVID-19 pandemic: a recent update. Curr. Pharmacol. Rep.6, 228–240 (2020).
    1. O’Brien MP, et al. Subcutaneous REGEN-COV antibody combination to prevent Covid-19. N. Engl. J. Med. 2021;385:1184–1195. doi: 10.1056/NEJMoa2109682.
    1. Chaudhary N, Weissman D, Whitehead KA. mRNA vaccines for infectious diseases: principles, delivery and clinical translation. Nat. Rev. Drug Discov. 2021;20:817–838. doi: 10.1038/s41573-021-00283-5.
    1. Aldrich C, et al. Proof-of-concept of a low-dose unmodified mRNA-based rabies vaccine formulated with lipid nanoparticles in human volunteers: A phase 1 trial. Vaccine. 2021;39:1310–1318. doi: 10.1016/j.vaccine.2020.12.070.
    1. Aliprantis AO, et al. A phase 1, randomized, placebo-controlled study to evaluate the safety and immunogenicity of an mRNA-based RSV prefusion F protein vaccine in healthy younger and older adults. Hum. Vaccin. Immunother. 2021;17:1248–1261. doi: 10.1080/21645515.2020.1829899.
    1. Feldman RA, et al. mRNA vaccines against H10N8 and H7N9 influenza viruses of pandemic potential are immunogenic and well tolerated in healthy adults in phase 1 randomized clinical trials. Vaccine. 2019;37:3326–3334. doi: 10.1016/j.vaccine.2019.04.074.
    1. Richner JM, et al. Modified mRNA vaccines protect against Zika virus infection. Cell. 2017;168:1114–1125. doi: 10.1016/j.cell.2017.02.017.
    1. Pardi N, et al. Administration of nucleoside-modified mRNA encoding broadly neutralizing antibody protects humanized mice from HIV-1 challenge. Nat. Commun. 2017;8:14630. doi: 10.1038/ncomms14630.
    1. Kose, N. et al. A lipid-encapsulated mRNA encoding a potently neutralizing human monoclonal antibody protects against chikungunya infection. Sci. Immunol.4, eaaw6647 (2019).
    1. Erasmus JH, et al. Intramuscular delivery of replicon RNA encoding ZIKV-117 human monoclonal antibody protects against Zika virus infection. Mol. Ther. Methods Clin. Dev. 2020;18:402–414. doi: 10.1016/j.omtm.2020.06.011.
    1. Gilchuk P, et al. Integrated pipeline for the accelerated discovery of antiviral antibody therapeutics. Nat. Biomed. Eng. 2020;4:1030–1043. doi: 10.1038/s41551-020-0594-x.
    1. Van Hoecke L, et al. mRNA encoding a bispecific single domain antibody construct protects against influenza a virus infection in mice. Mol. Ther. Nucleic Acids. 2020;20:777–787. doi: 10.1016/j.omtn.2020.04.015.
    1. Zhu L, et al. Double lock of a potent human therapeutic monoclonal antibody against SARS-CoV-2. Natl. Sci. Rev. 2021;8:nwaa297. doi: 10.1093/nsr/nwaa297.
    1. Zhang NN, et al. A thermostable mRNA vaccine against COVID-19. Cell. 2020;182:1271–1283.e1216. doi: 10.1016/j.cell.2020.07.024.
    1. Zhang NN, et al. A thermostable mRNA vaccine against COVID-19. Cell. 2020;182:1271–1283.e1216. doi: 10.1016/j.cell.2020.07.024.
    1. Sun S, et al. Characterization and structural basis of a lethal mouse-adapted SARS-CoV-2. Nat. Commun. 2021;12:5654. doi: 10.1038/s41467-021-25903-x.
    1. Chen Q, et al. Transient acquisition of cross-species infectivity during the evolution of SARS-CoV-2. Natl. Sci. Rev. 2021;8:nwab167. doi: 10.1093/nsr/nwab167.
    1. Sia SF, et al. Pathogenesis and transmission of SARS-CoV-2 in golden hamsters. Nature. 2020;583:834–838. doi: 10.1038/s41586-020-2342-5.
    1. Taylor PC, et al. Neutralizing monoclonal antibodies for treatment of COVID-19. Nat. Rev. Immunol. 2021;21:382–393. doi: 10.1038/s41577-021-00542-x.
    1. Hurt, A. C. & Wheatley, A. K. Neutralizing antibody therapeutics for COVID-19. Viruses13, 628 (2021).
    1. Thran M, et al. mRNA mediates passive vaccination against infectious agents, toxins, and tumors. EMBO Mol. Med. 2017;9:1434–1447. doi: 10.15252/emmm.201707678.
    1. Wang Z, et al. mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants. Nature. 2021;592:616–622. doi: 10.1038/s41586-021-03324-6.
    1. Chen RE, et al. In vivo monoclonal antibody efficacy against SARS-CoV-2 variant strains. Nature. 2021;596:103–108. doi: 10.1038/s41586-021-03720-y.

Source: PubMed

3
Se inscrever