RT Journal Article SR Electronic T1 Broad immunity to SARS-CoV-2 variants of concern mediated by a SARS-CoV-2 receptor-binding domain protein vaccine JF medRxiv FD Cold Spring Harbor Laboratory Press SP 2022.08.05.22278425 DO 10.1101/2022.08.05.22278425 A1 Georgia Deliyannis A1 Nicholas A. Gherardin A1 Chinn Yi Wong A1 Samantha L. Grimley A1 James P. Cooney A1 Samuel Redmond A1 Paula Ellenberg A1 Kathryn Davidson A1 Francesca L. Mordant A1 Tim Smith A1 Marianne Gillard A1 Ester Lopez A1 Julie McAuley A1 Chee Wah Tan A1 Jing Wang A1 Weiguang Zeng A1 Mason Littlejohn A1 Runhong Zhou A1 Jasper Fuk-Woo Chan A1 Zhi-wei Chen A1 Airn E. Hartwig A1 Richard Bowen A1 Jason M. Mackenzie A1 Elizabeth Vincan A1 Joseph Torresi A1 Katherine Kedzierska A1 Colin W. Pouton A1 Tom Gordon A1 Lin-fa Wang A1 Stephen J. Kent A1 Adam K. Wheatley A1 Sharon R. Lewin A1 Kanta Subbarao A1 Amy Chung A1 Marc Pellegrini A1 Trent Munro A1 Terry Nolan A1 Steven Rockman A1 David C. Jackson A1 Damian F.J. Purcell A1 Dale I. Godfrey YR 2022 UL http://medrxiv.org/content/early/2022/08/09/2022.08.05.22278425.abstract AB The SARS-CoV-2 global pandemic has fuelled the generation of vaccines at an unprecedented pace and scale. However, many challenges remain, including: the emergence of vaccine-resistant mutant viruses, vaccine stability during storage and transport, waning vaccine-induced immunity, and concerns about infrequent adverse events associated with existing vaccines. Here, we report on a protein subunit vaccine comprising the receptor-binding domain (RBD) of the ancestral SARS-CoV-2 spike protein, dimerised with an immunoglobulin IgG1 Fc domain. These were tested in conjunction with three different adjuvants: a TLR2 agonist R4-Pam2Cys, an NKT cell agonist glycolipid α-Galactosylceramide, or MF59® squalene oil-in-water adjuvant. Each formulation drove strong neutralising antibody (nAb) responses and provided durable and highly protective immunity against lower and upper airway infection in mouse models of COVID-19. We have also developed an RBD-human IgG1 Fc vaccine with an RBD sequence of the highly immuno-evasive beta variant (N501Y, E484K, K417N). This ‘beta variant’ RBD vaccine, combined with MF59® adjuvant, induced strong protection in mice against the beta strain as well as the ancestral strain. Furthermore, when used as a third dose booster vaccine following priming with whole spike vaccine, anti-sera from beta-RBD-Fc immunised mice increased titres of nAb against other variants including alpha, delta, delta+, gamma, lambda, mu, and omicron BA.1 and BA.2. These results demonstrated that an RBD-Fc protein subunit/MF59® adjuvanted vaccine can induce high levels of broad nAbs, including when used as a booster following prior immunisation of mice with whole ancestral-strain Spike vaccines. This vaccine platform offers a potential approach to augment some of the currently approved vaccines in the face of emerging variants of concern, and it has now entered a phase I clinical trial.Competing Interest StatementTwo provisional patents covering the RBD-Fc vaccines described in this study, and underlying technology, have been submitted through The University of Melbourne. CWT and L-FW are co-inventors of a patent on the surrogate virus neutralization test (sVNT) platform.Funding StatementThis work was supported by grants from the Medical Research Future Fund (MRFF) Awards (2005544, 2002073, 2002132), The Jack Ma Foundation, and National Health and Medical Research Council of Australia (NHMRC; 1113293, 2002317 and 1116530). GD was supported by philanthropic funds from IFM, DIG was supported by an NHMRC Senior Principal Research Fellowship (1117766) and an NHMRC Investigator Award (2008913). KK was supported by the NHMRC Leadership Investigator Grant (1173871), KS also received support from the A2 Milk Company. The Melbourne WHO Collaborating Centre for Reference and Research on Influenza is supported by the Australian Government Department of Health. The work at Duke-NUS is supported by grant from Singapore National Medical Research Council (MOH-COVID19RF-003).Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:The University of Melbourne Human Research Ethics Committee (2021-21198-15398-3, 2056689), gave ethical approval for the human sample studies in this work.I confirm that all necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived, and that any patient/participant/sample identifiers included were not known to anyone (e.g., hospital staff, patients or participants themselves) outside the research group so cannot be used to identify individuals.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesAll data produced in the present study are available upon reasonable request to the authors