Evaluation of the mRNA-1273 Vaccine against SARS-CoV-2 in Nonhuman Primates

Kizzmekia S Corbett, Barbara Flynn, Kathryn E Foulds, Joseph R Francica, Seyhan Boyoglu-Barnum, Anne P Werner, Britta Flach, Sarah O'Connell, Kevin W Bock, Mahnaz Minai, Bianca M Nagata, Hanne Andersen, David R Martinez, Amy T Noe, Naomi Douek, Mitzi M Donaldson, Nadesh N Nji, Gabriela S Alvarado, Darin K Edwards, Dillon R Flebbe, Evan Lamb, Nicole A Doria-Rose, Bob C Lin, Mark K Louder, Sijy O'Dell, Stephen D Schmidt, Emily Phung, Lauren A Chang, Christina Yap, John-Paul M Todd, Laurent Pessaint, Alex Van Ry, Shanai Browne, Jack Greenhouse, Tammy Putman-Taylor, Amanda Strasbaugh, Tracey-Ann Campbell, Anthony Cook, Alan Dodson, Katelyn Steingrebe, Wei Shi, Yi Zhang, Olubukola M Abiona, Lingshu Wang, Amarendra Pegu, Eun Sung Yang, Kwanyee Leung, Tongqing Zhou, I-Ting Teng, Alicia Widge, Ingelise Gordon, Laura Novik, Rebecca A Gillespie, Rebecca J Loomis, Juan I Moliva, Guillaume Stewart-Jones, Sunny Himansu, Wing-Pui Kong, Martha C Nason, Kaitlyn M Morabito, Tracy J Ruckwardt, Julie E Ledgerwood, Martin R Gaudinski, Peter D Kwong, John R Mascola, Andrea Carfi, Mark G Lewis, Ralph S Baric, Adrian McDermott, Ian N Moore, Nancy J Sullivan, Mario Roederer, Robert A Seder, Barney S Graham, Kizzmekia S Corbett, Barbara Flynn, Kathryn E Foulds, Joseph R Francica, Seyhan Boyoglu-Barnum, Anne P Werner, Britta Flach, Sarah O'Connell, Kevin W Bock, Mahnaz Minai, Bianca M Nagata, Hanne Andersen, David R Martinez, Amy T Noe, Naomi Douek, Mitzi M Donaldson, Nadesh N Nji, Gabriela S Alvarado, Darin K Edwards, Dillon R Flebbe, Evan Lamb, Nicole A Doria-Rose, Bob C Lin, Mark K Louder, Sijy O'Dell, Stephen D Schmidt, Emily Phung, Lauren A Chang, Christina Yap, John-Paul M Todd, Laurent Pessaint, Alex Van Ry, Shanai Browne, Jack Greenhouse, Tammy Putman-Taylor, Amanda Strasbaugh, Tracey-Ann Campbell, Anthony Cook, Alan Dodson, Katelyn Steingrebe, Wei Shi, Yi Zhang, Olubukola M Abiona, Lingshu Wang, Amarendra Pegu, Eun Sung Yang, Kwanyee Leung, Tongqing Zhou, I-Ting Teng, Alicia Widge, Ingelise Gordon, Laura Novik, Rebecca A Gillespie, Rebecca J Loomis, Juan I Moliva, Guillaume Stewart-Jones, Sunny Himansu, Wing-Pui Kong, Martha C Nason, Kaitlyn M Morabito, Tracy J Ruckwardt, Julie E Ledgerwood, Martin R Gaudinski, Peter D Kwong, John R Mascola, Andrea Carfi, Mark G Lewis, Ralph S Baric, Adrian McDermott, Ian N Moore, Nancy J Sullivan, Mario Roederer, Robert A Seder, Barney S Graham

Abstract

Background: Vaccines to prevent coronavirus disease 2019 (Covid-19) are urgently needed. The effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines on viral replication in both upper and lower airways is important to evaluate in nonhuman primates.

Methods: Nonhuman primates received 10 or 100 μg of mRNA-1273, a vaccine encoding the prefusion-stabilized spike protein of SARS-CoV-2, or no vaccine. Antibody and T-cell responses were assessed before upper- and lower-airway challenge with SARS-CoV-2. Active viral replication and viral genomes in bronchoalveolar-lavage (BAL) fluid and nasal swab specimens were assessed by polymerase chain reaction, and histopathological analysis and viral quantification were performed on lung-tissue specimens.

Results: The mRNA-1273 vaccine candidate induced antibody levels exceeding those in human convalescent-phase serum, with live-virus reciprocal 50% inhibitory dilution (ID50) geometric mean titers of 501 in the 10-μg dose group and 3481 in the 100-μg dose group. Vaccination induced type 1 helper T-cell (Th1)-biased CD4 T-cell responses and low or undetectable Th2 or CD8 T-cell responses. Viral replication was not detectable in BAL fluid by day 2 after challenge in seven of eight animals in both vaccinated groups. No viral replication was detectable in the nose of any of the eight animals in the 100-μg dose group by day 2 after challenge, and limited inflammation or detectable viral genome or antigen was noted in lungs of animals in either vaccine group.

Conclusions: Vaccination of nonhuman primates with mRNA-1273 induced robust SARS-CoV-2 neutralizing activity, rapid protection in the upper and lower airways, and no pathologic changes in the lung. (Funded by the National Institutes of Health and others.).

Copyright © 2020 Massachusetts Medical Society.

Figures

Figure 1. Antibody Responses after mRNA-1273 Vaccination…
Figure 1. Antibody Responses after mRNA-1273 Vaccination in Rhesus Macaques.
Animals were administered phosphate-buffered saline (PBS) as a control or 10 μg or 100 μg of mRNA-1273. Serum specimens were assessed for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) S-specific IgG by enzyme-linked immunosorbent assay (ELISA) (Panel A) and SARS-CoV-2 pseudovirus neutralization (Panel B) at all time points after the first and second vaccinations. Data in Panel A are the area under the curve (AUC) and indicate the amount of IgG binding to S-2P over time, and data in Panel B are the reciprocal 50% inhibitory dilution (ID50). Faint lines in Panels A and B represent individual animals, and bold lines represent the geometric mean titer for each group. S-specific IgG (Panel C), pseudovirus neutralization (Panel D), inhibition of angiotensin-converting enzyme 2 (ACE2) binding to the receptor-binding domain (RBD) (Panel E), and live-virus neutralization by NanoLuc reporter assay (Promega) (Panel F) were assessed at 4 weeks after the second vaccination, immediately before challenge. Results were compared with the antibody responses in a panel of human convalescent-phase serum specimens (Conv.) (42 specimens in Panels C, D, and E and 26 specimens in Panel F). In Panel E, the amount of signal emitted in wells containing no specimen was used as the maximal binding response against which each factor reduction was measured. In the box-and-whisker plots, the horizontal line indicates the median, the top and bottom of the box the interquartile range, and the whiskers the range. Symbols represent individual animals and overlap with one another for equal values where constrained. Dashed lines indicate the assay limit of detection.
Figure 2. T-Cell Responses after mRNA-1273 Vaccination…
Figure 2. T-Cell Responses after mRNA-1273 Vaccination in Rhesus Macaques.
Intracellular staining was performed on peripheral blood mononuclear cells at 8 weeks, immediately before challenge, to assess T-cell responses to the SARS-CoV-2 S1 peptide pool. Panel A shows type 1 helper T-cell (Th1) responses (interferon-γ, interleukin-2, or tumor necrosis factor α), Panel B Th2 responses (interleukin-4 or 13), Panel C CD40L up-regulation, and Panel D interleukin-21 from peripheral follicular helper T (Tfh) cells (central memory CXCR5+PD-1+ICOS+ CD4 T cells). Positivity with respect to intracellular cytokine responses was determined with the MIMOSA algorithm; numbers of animals positive and total numbers of animals are shown as fractions below each group. In the box-and-whisker plots, the horizontal line indicates the median, the top and bottom of the box the interquartile range, and the whiskers the range. Open symbols represent animals with a probable nonresponse, and solid symbols represent animals with a probable response. Dashed lines are used to highlight 0.0%.
Figure 3. Efficacy of mRNA-1273 against Upper…
Figure 3. Efficacy of mRNA-1273 against Upper and Lower Respiratory Viral Replication.
Bronchoalveolar-lavage (BAL) fluid (Panel A) and nasal swab (Panel B) specimens were obtained on days 1, 2, 4, and 7 after challenge, where applicable, and viral replication was assessed by analysis of SARS-CoV-2 subgenomic RNA. In the box-and-whisker plots, the horizontal line indicates the median, the top and bottom of the box the interquartile range, and the whiskers the range. Symbols represent individual animals and overlap with one another for equal values where constrained. Dashed lines indicate the assay limit of detection.
Figure 4. Lung Histopathological Analysis and Viral…
Figure 4. Lung Histopathological Analysis and Viral Detection 7 Days after Challenge in mRNA-1273–Vaccinated Rhesus Macaques.
Seven days after challenge, lungs were harvested from two animals per group for histopathological analysis and assessment of evidence of viral infection; representative images taken at different degrees of magnification are shown for localization of virus by chromogenic in situ hybridization (CISH) and SARS-CoV-2 immunohistochemical analysis (IHC) in serial tissue sections. The images in Panel B are shown at twice the magnification of the images in Panel A.

References

    1. Cucinotta D, Vanelli M. WHO declares COVID-19 a pandemic. Acta Biomed 2020;91:157-160.
    1. Callaway E, Cyranoski D, Mallapaty S, Stoye E, Tollefson J. The coronavirus pandemic in five powerful charts. Nature 2020;579:482-483.
    1. Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 2020;181(2):271.e8-280.e8.
    1. Amanat F, Krammer F. SARS-CoV-2 vaccines: status report. Immunity 2020;52:583-589.
    1. Mulligan MJ, Lyke KE, Kitchin N, et al. Phase 1/2 study to describe the safety and immunogenicity of a COVID-19 RNA vaccine candidate (BNT162b1) in adults 18 to 55 years of age: interim report. July 1, 2020. (). preprint.
    1. Corbett KS, Edwards D, Leist SR, et al. SARS-CoV-2 mRNA vaccine development enabled by prototype pathogen preparedness. June 11, 2020. (). preprint.
    1. Yu J, Tostanoski LH, Peter L, et al. DNA vaccine protection against SARS-CoV-2 in rhesus macaques. Science 2020. May 20 (Epub ahead of print).
    1. Smith TRF, Patel A, Ramos S, et al. Immunogenicity of a DNA vaccine candidate for COVID-19. Nat Commun 2020;11:2601-2601.
    1. Zhu F-C, Li Y-H, Guan X-H, et al. Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: a dose-escalation, open-label, non-randomised, first-in-human trial. Lancet 2020;395:1845-1854.
    1. van Doremalen N, Lambe T, Spencer A, et al. ChAdOx1 nCoV-19 vaccination prevents SARS-CoV-2 pneumonia in rhesus macaques. May 13, 2020. (). preprint.
    1. Gao Q, Bao L, Mao H, et al. Development of an inactivated vaccine candidate for SARS-CoV-2. Science 2020;369:77-81.
    1. Wang H, Zhang Y, Huang B, et al. Development of an inactivated vaccine candidate, BBIBP-CorV, with potent protection against SARS-CoV-2. Cell. June 6, 2020. ().
    1. Chen W-H, Strych U, Hotez PJ, Bottazzi ME. The SARS-CoV-2 vaccine pipeline: an overview. Curr Trop Med Rep 2020. March 3 (Epub ahead of print).
    1. Munster VJ, Feldmann F, Williamson BN, et al. Respiratory disease and virus shedding in rhesus macaques inoculated with SARS-CoV-2. March 21, 2020. (). preprint.
    1. Bahl K, Senn JJ, Yuzhakov O, et al. Preclinical and clinical demonstration of immunogenicity by mRNA vaccines against H10N8 and H7N9 influenza viruses. Mol Ther 2017;25:1316-1327.
    1. Jackson LA, Anderson EJ, Rouphael NG, et al. An mRNA vaccine against SARS-CoV-2 — preliminary report. N Engl J Med. DOI: 10.1056/NEJMoa2022483.
    1. Hassett KJ, Benenato KE, Jacquinet E, et al. Optimization of lipid nanoparticles for intramuscular administration of mRNA vaccines. Mol Ther Nucleic Acids 2019;15:1-11.
    1. Wölfel R, Corman VM, Guggemos W, et al. Virological assessment of hospitalized patients with COVID-2019. Nature 2020;581:465-469.
    1. Wang F, Flanagan J, Su N, et al. RNAscope: a novel in situ RNA analysis platform for formalin-fixed, paraffin-embedded tissues. J Mol Diagn 2012;14:22-29.
    1. Hou YJ, Okuda K, Edwards CE, et al. SARS-CoV-2 reverse genetics reveals a variable infection gradient in the respiratory tract. Cell 2020;182(2):429-446.e14.
    1. Scobey T, Yount BL, Sims AC, et al. Reverse genetics with a full-length infectious cDNA of the Middle East respiratory syndrome coronavirus. Proc Natl Acad Sci U S A 2013;110:16157-16162.
    1. Yount B, Curtis KM, Fritz EA, et al. Reverse genetics with a full-length infectious cDNA of severe acute respiratory syndrome coronavirus. Proc Natl Acad Sci USA 2003;100:12995-13000.
    1. Donaldson MM, Kao S-F, Foulds KE. OMIP-052: an 18-color panel for measuring Th1, Th2, Th17, and Tfh responses in rhesus macaques. Cytometry A 2019;95:261-263.
    1. Finak G, McDavid A, Chattopadhyay P, et al. Mixture models for single-cell assays with applications to vaccine studies. Biostatistics 2014;15:87-101.
    1. Pallesen J, Wang N, Corbett KS, et al. Immunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigen. Proc Natl Acad Sci U S A 2017;114:E7348-E7357.
    1. Wrapp D, Wang N, Corbett KS, et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 2020;367:1260-1263.
    1. Wang L, Shi W, Chappell JD, et al. Importance of neutralizing monoclonal antibodies targeting multiple antigenic sites on the Middle East respiratory syndrome coronavirus spike glycoprotein to avoid neutralization escape. J Virol 2018;92(10):e02002-17-e02002-17.
    1. Wang N, Rosen O, Wang L, et al. Structural definition of a neutralization-sensitive epitope on the MERS-CoV S1-NTD. Cell Rep 2019;28(13):3395.e6-3405.e6.
    1. Chen Y, Lu S, Jia H, et al. A novel neutralizing monoclonal antibody targeting the N-terminal domain of the MERS-CoV spike protein. Emerg Microbes Infect 2017;6(5):e37-e37.
    1. Rogers TF, Zhao F, Huang D, et al. Isolation of potent SARS-CoV-2 neutralizing antibodies and protection from disease in a small animal model. Science 2020. June 15 (Epub ahead of print).
    1. Shi R, Shan C, Duan X, et al. A human neutralizing antibody targets the receptor-binding site of SARS-CoV-2. Nature 2020. May 26 (Epub ahead of print).
    1. Wang W, Wang H, Deng Y, et al. Characterization of anti-MERS-CoV antibodies against various recombinant structural antigens of MERS-CoV in an imported case in China. Emerg Microbes Infect 2016;5(11):e113-e113.
    1. Widjaja I, Wang C, van Haperen R, et al. Towards a solution to MERS: protective human monoclonal antibodies targeting different domains and functions of the MERS-coronavirus spike glycoprotein. Emerg Microbes Infect 2019;8:516-530.
    1. Neidleman J, Luo X, Frouard J, et al. SARS-CoV-2-specific T cells exhibit unique features characterized by robust helper function, lack of terminal differentiation, and high proliferative potential. June 8, 2020. (). preprint.
    1. Weiskopf D, Schmitz KS, Raadsen MP, et al. Phenotype and kinetics of SARS-CoV-2-specific T cells in COVID-19 patients with acute respiratory distress syndrome. Sci Immunol 2020;5:eabd2071-eabd2071.
    1. Meckiff BJ, Ramírez-Suástegui C, Fajardo V, et al. Single-cell transcriptomic analysis of SARS-CoV-2 reactive CD4 + T cells. June 13, 2020. (). preprint.
    1. Grifoni A, Weiskopf D, Ramirez SI, et al. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell 2020;181(7):1489.e15-1501.e15.
    1. Peng Y, Mentzer AJ, Liu G, et al. Broad and strong memory CD4+ and CD8+ T cells induced by SARS-CoV-2 in UK convalescent COVID-19 patients. June 8, 2020. (). preprint.
    1. Sekine T, Perez-Potti A, Rivera-Ballesteros O, et al. Robust T cell immunity in convalescent individuals with asymptomatic or mild COVID-19. June 29, 2020. (). preprint.
    1. Pardi N, Hogan MJ, Naradikian MS, et al. Nucleoside-modified mRNA vaccines induce potent T follicular helper and germinal center B cell responses. J Exp Med 2018;215:1571-1588.
    1. Zou L, Ruan F, Huang M, et al. SARS-CoV-2 viral load in upper respiratory specimens of infected patients. N Engl J Med 2020;382:1177-1179.
    1. Chandrashekar A, Liu J, Martinot AJ, et al. SARS-CoV-2 infection protects against rechallenge in rhesus macaques. Science 2020. May 20 (Epub ahead of print).
    1. Rockx B, Kuiken T, Herfst S, et al. Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model. Science 2020;368:1012-1015.
    1. Edridge AWD, Kaczorowska JM, Hoste ACR, et al. Coronavirus protective immunity is short-lasting. June 16, 2020. (). preprint.
    1. Long Q-X, Tang X-J, Shi Q-L, et al. Clinical and immunological assessment of asymptomatic SARS-CoV-2 infections. Nat Med 2020. June 18 (Epub ahead of print).
    1. Crank MC, Ruckwardt TJ, Chen M, et al. A proof of concept for structure-based vaccine design targeting RSV in humans. Science 2019;365:505-509.
    1. Graham BS, Gilman MSA, McLellan JS. Structure-based vaccine antigen design. Annu Rev Med 2019;70:91-104.
    1. Stewart-Jones GBE, Chuang G-Y, Xu K, et al. Structure-based design of a quadrivalent fusion glycoprotein vaccine for human parainfluenza virus types 1-4. Proc Natl Acad Sci U S A 2018;115:12265-12270.
    1. Loomis RJ, Stewart-Jones GBE, Tsybovsky Y, et al. Structure-based design of Nipah virus vaccines: a generalizable approach to paramyxovirus immunogen development. Front Immunol 2020;11:842-842.
    1. Sanders RW, Vesanen M, Schuelke N, et al. Stabilization of the soluble, cleaved, trimeric form of the envelope glycoprotein complex of human immunodeficiency virus type 1. J Virol 2002;76:8875-8889.
    1. Chuang GY, Geng H, Pancera M, et al. Structure-based design of a soluble prefusion-closed HIV-1 Env trimer with reduced CD4 affinity and improved immunogenicity. J Virol 2017;91(10):e02268-16-e02268-16.
    1. Killikelly AM, Kanekiyo M, Graham BS. Pre-fusion F is absent on the surface of formalin-inactivated respiratory syncytial virus. Sci Rep 2016;6:34108-34108.
    1. Karikó K, Muramatsu H, Welsh FA, et al. Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stability. Mol Ther 2008;16:1833-1840.
    1. Graham BS. Rapid COVID-19 vaccine development. Science 2020;368:945-946.

Source: PubMed

3
Sottoscrivi