TY - JOUR T1 - mRNA vaccine-induced IgA protect against SARS-CoV-2 breakthrough infection JF - medRxiv DO - 10.1101/2021.08.01.21261297 SP - 2021.08.01.21261297 AU - Salma Sheikh-Mohamed AU - Baweleta Isho AU - Gary Y.C. Chao AU - Michelle Zuo AU - Carmit Cohen AU - Yaniv Lustig AU - George R. Nahass AU - Rachel E. Salomon-Shulman AU - Grace Blacker AU - Mahya Fazel-Zarandi AU - Bhavisha Rathod AU - Karen Colwill AU - Alainna Jamal AU - Zhijie Li AU - Keelia Quin de Launay AU - Alyson Takaoka AU - Julia Garnham-Takaoka AU - Anjali Patel AU - Christine Fahim AU - Aimee Paterson AU - Angel Xinliu Li AU - Nazrana Haq AU - Shiva Barati AU - Lois Gilbert AU - Karen Green AU - Mohammad Mozafarihashjin AU - Philip Samaan AU - Patrick Budylowski AU - Walter L. Siqueira AU - Samira Mubareka AU - Mario Ostrowski AU - James M. Rini AU - Olga L. Rojas AU - Irving L. Weissman AU - Michal Caspi Tal AU - Allison McGeer AU - Gili Regev-Yochay AU - Sharon Straus AU - Anne-Claude Gingras AU - Jennifer L. Gommerman Y1 - 2021/01/01 UR - http://medrxiv.org/content/early/2021/12/15/2021.08.01.21261297.abstract N2 - Background Although SARS-CoV-2 infects the upper respiratory tract, we know little about the amount, type, and kinetics of antibodies (Ab) generated at this site in response to intramuscular COVID-19 vaccination, and whether these Ab protect against breakthrough infections.Methods We collected longitudinal serum and saliva samples from participants receiving two doses of mRNA COVID-19 vaccines over a 6-month period and measured the relative level of anti-Spike and anti-Receptor Binding Domain (RBD) IgM, IgG and IgA Ab and associated secretory component. We also compared the levels of anti-Spike Ab at 2-4 weeks post dose 2 in participants who subsequently experienced a breakthrough infection in two separate prospective cohorts.Results We detected anti-Spike/RBD IgG and IgA and associated secretory component in the saliva of most participants receiving 1 dose of mRNA vaccine. At 6 months post-dose 2, salivary anti-Spike/RBD IgG levels were still detected but greatly diminished. In contrast, anti-Spike/RBD IgA levels dropped much earlier – between dose 1 and dose 2 - with only 30% of participants retaining IgA levels after dose 2. Analysis of participants who subsequently experienced a breakthrough infection revealed that serum IgA but not IgG anti-Spike antibodies were lower in breakthrough infected cases versus uninfected control participants.Conclusions COVID-19 mRNA vaccination induces a local anti-Spike/RBD IgA response with variable persistence. Participants who experience a breakthrough infection have lower levels of anti-Spike IgA at 2-4 weeks post dose 2 compared to controls. These data emphasize the importance vaccine elicited IgA in preventing breakthrough infections.One-Sentence Summary Our study delves into whether intra-muscular mRNA/mRNA vaccination regimes confer a local IgA response in the oral cavity and whether this response is associated with protection against breakthrough infection.Competing Interest StatementThe authors have declared no competing interest.Funding StatementThis work was supported by an Ontario Together province of Ontario grant to JG and ACG and a Foundation grant from the Canadian Institutes of Health Research to JG (Fund #15992). Funding for the LTCH cohort was provided through a Canada COVID-19 Immunity Task force grant (to SS, AM, MO, ACG and JG). Funding for initial development of the assays in the Gingras lab was provided through generous donations from the Royal Bank of Canada (RBC) and the Krembil Foundation to the Sinai Health System Foundation. The robotics equipment used is housed in the Network Biology Collaborative Centre at the Lunenfeld-Tanenbaum Research Institute, a facility supported by Canada Foundation for Innovation funding, by the Ontarian Government and by Genome Canada and Ontario Genomics (OGI-139).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 Mount Sinai Hospital Research Ethics Board (REB) granted approval for recruiting LTCH participants located in Toronto for blood and saliva collection and for conducting serum ELISAs at the Lunenfeld-Tanenbaum Research Institute (study number: 20-0339-E). The University of Toronto REB granted approval for subject recruitment to collect blood and saliva samples and for conducting saliva ELISAs (study number: 23901). The University of Saskatchewan REB granted approval for saliva sample collection during the pre-COVID era (study number: BIO-USask-1579). The Sheba Medical Center REB granted approval for subject recruitment to collect blood and for conducting ELISAs (study number: 8008-20-SMC). Cohorts are further described in Tables S1-S6 and supplemental methods in the Appendix.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.YesThe data will be available upon reasonable request. ER -