mRNA vaccine-induced IgA protect against SARS-CoV-2 breakthrough infection ========================================================================== * Salma Sheikh-Mohamed * Baweleta Isho * Gary Y.C. Chao * Michelle Zuo * Carmit Cohen * Yaniv Lustig * George R. Nahass * Rachel E. Salomon-Shulman * Grace Blacker * Mahya Fazel-Zarandi * Bhavisha Rathod * Karen Colwill * Alainna Jamal * Zhijie Li * Keelia Quin de Launay * Alyson Takaoka * Julia Garnham-Takaoka * Anjali Patel * Christine Fahim * Aimee Paterson * Angel Xinliu Li * Nazrana Haq * Shiva Barati * Lois Gilbert * Karen Green * Mohammad Mozafarihashjin * Philip Samaan * Patrick Budylowski * Walter L. Siqueira * Samira Mubareka * Mario Ostrowski * James M. Rini * Olga L. Rojas * Irving L. Weissman * Michal Caspi Tal * Allison McGeer * Gili Regev-Yochay * Sharon Straus * Anne-Claude Gingras * Jennifer L. Gommerman ## Abstract **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. Key Words * SARS-CoV-2 * mucosal * intramuscular * vaccination * mRNA * antibodies * secretory component ## Introduction SARS-CoV-2 is a novel and highly contagious respiratory virus that has quickly spread across the globe. The virus uses a protein called Spike and its associated receptor binding domain (RBD) to interact with angiotensin converting enzyme 2 (ACE2) on the surface of epithelial cells in the upper respiratory tract1. SARS-CoV-2 is mainly transmitted via inhaled aerosols, making the immune response in the oral and nasal mucosa an important first line of defense2. Saliva is an important biofluid that can provide information about the mucosal antibody (Ab) response to SARS-CoV-23. Indeed, the salivary glands themselves express ACE2 and harbor a significant population of IgA-producing plasma cells3,4. Secretory IgA (sIgA) in the saliva exists as IgA dimers that are associated with the secretory component, a proteolytic cleavage product which remains bound to IgA after it is transported across epithelial cells via the polymeric Ig receptor (pIgR)5. Secretory polymeric IgA has been shown to have potent neutralizing activity against SARS-CoV-2 *in vitro*6. We and others have shown that IgM, IgG and IgA Ab against the SARS-CoV-2 Spike and RBD proteins are readily detected in the saliva of COVID-19 acute and convalescent patients3,7. Whether COVID-19 vaccines delivered through the parenteral intramuscular route (i.m.) generate a similar salivary antibody response is unclear, and the nature and kinetics of this response are ill-characterized. To address this gap, we measured SARS-CoV-2-specific Ab in saliva samples from participants who had received either BNT162b2 or mRNA-1273 vaccinations. We also determined whether levels of vaccine induced SARS-CoV-2 specific IgG or IgA differed in people who subsequently experienced a breakthrough infection. Collectively, our data show that IgA is induced in some people vaccinated against COVID-19, and that IgA may play an important role in protection against breakthrough infection. ## Materials and Methods ### Study approvals 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. ### Enzyme-linked immunosorbent assays for detecting antigen-specific IgG, IgA, and IgM in serum We used a previously published method for detection of saliva and serum anti-SARS-CoV-2 antibodies3. Details for this assay are in the methods in the supplemental methods in the Appendix. ### Enzyme-linked immunosorbent assays for detecting anti-Spike and anti-RBD IgA, IgG and IgM in saliva Saliva collection, antigen production methods, detailed protocol and analytical approach can be found in the methods in the supplemental methods in the Appendix. Here we used saliva from COVID-19 acute and convalescent patients, analysed as in3. COVID-19 saliva positive controls and pre-COVID-19 saliva negative controls were used as comparators. ### ELISA for detection of secretory component associated anti-Spike/RBD antibodies Secretory chain associated antibodies were detected by modifying our saliva Spike/RBD ELISA using an HRP-conjugated Goat anti-human secretory component detection reagent at a dilution of 1/750 from Nordic MUBio (Cat# GAHu/SC/PO). ## Results ### Analysis of anti-Spike and anti-RBD antibodies in saliva from participants receiving COVID-19 mRNA vaccines We first compared saliva from long-term care home (LTCH) workers that received either BNT162b2 or mRNA-1273 (Fig. 1 and Table S1) with pooled negative controls used to establish a cut-off (Tables S2a-b), and COVID-19 acute and convalescent patients as positive controls (Table S2c). We expressed the data as a percentage relative to a pooled positive control that was present in each plate (see supplemental methods in the Appendix). We found that 11%, 94% and 41% of participants were positive for anti-Spike IgM, IgG and IgA and 22%, 93% and 20% of participants were positive for anti-RBD IgM, IgG and IgA Ab (Fig. 2 A-F). Furthermore, as we observed before in COVID-19 recovered patients3, levels of salivary anti-Spike/RBD Ab correlated well with anti-Spike/RBD Ab in the serum (Fig. S1). In multivariable analysis, age and prior SARS-CoV-2 infection were independently associated with the salivary anti-Spike IgA response (Table S3a). In contrast, male sex had a negative independent association with the salivary anti-Spike IgG response (Table S3b) as has been observed before for COVID-19 and other vaccines8,9. Lastly, prior SARS-CoV-2 infection and time since vaccination were independently associated with higher and lower serum anti-RBD IgA levels, respectively (Table S3c). ![Fig. 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/12/15/2021.08.01.21261297/F1.medium.gif) [Fig. 1:](http://medrxiv.org/content/early/2021/12/15/2021.08.01.21261297/F1) Fig. 1: Study Design. Two observational cohort (LTCH staff, MSB) and two prospective cohorts (LTCH Residents, Sheba Medical Center HCW) were integrated into our overall study of mRNA/mRNA vaccinated subjects. ![Fig 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/12/15/2021.08.01.21261297/F2.medium.gif) [Fig 2.](http://medrxiv.org/content/early/2021/12/15/2021.08.01.21261297/F2) Fig 2. Analysis of anti-Spike and anti-RBD antibodies in saliva from participants receiving COVID-19 mRNA vaccines. (**A-F**) Anti-Spike/RBD salivary Ab were measured in vaccinated LTCH participants after two-doses of either BNT162b2 or mRNA-1273. Levels were compared to COVID-19 controls and individually run negative controls. The positive cutoff (dotted line) was calculated as 2 standard deviations above the mean of a pool of negative control samples (n=51) for each individual assay. (**G-L**) Anti-Spike/RBD salivary Ab were also measured in vaccinated participants that were followed at sequential timepoints before and after dose 1 and dose 2 of BNT162b2 (MSB, n=27 with † = n=11 participants at this timepoint). All data is expressed as a percentage of the pooled positive plate control (see Methods). Solid black bars denote the median for each cohort. Mann-Whitney U test was used to calculate significance, where ns=not significant, *=p<0.05, **=p<0.01, \***|=p<0.001 and \**\*|\*=p<0.0001. As of June 2021, although most LTCH workers had been fully vaccinated, significant sectors of the Canadian population had only been administered a single dose of a COVID-19 vaccine because the interval between dose 1 vs dose 2 was extended as a dose sparing measure. Thus, we wished to ascertain if salivary Ab could be detected after a single dose of a COVID-19 mRNA vaccine, and how long these Ab would persist. We therefore collected samples from a second cohort of healthy adults that were followed longitudinally (Medical Sciences Building cohort – MSB; Fig. 1 and Table S4). These participants received 1 dose of BNT1162b2 and a second dose of BNT1162b2 3 months later, with samples taken at baseline, 2 weeks post-dose 1, 3 months post-dose 1, and 2-weeks post-dose 2. For the entire sampling period, of those participants that did not show evidence of prior SARS-CoV-2 infection IgM levels were higher than baseline only for anti-Spike post-dose 2, and anti-RBD at 2 weeks post-dose 1. Focusing therefore on IgG and IgA, we observed that 97% and 93% of participants were positive for anti-Spike IgG and IgA, and 52% and 41% were positive for anti-RBD IgG and IgA Ab in their saliva 2 weeks post-dose 1 (Fig. 2G-L). Of note, three months after dose 1, the median level of salivary anti-Spike/RBD Ab had diminished nearly to baseline. Following administration of a second dose of mRNA, while antigen-specific IgG levels recovered upon administration of dose 2 as expected, a second dose did not augment antigen-specific salivary IgA levels in most subjects (Fig. 2I-J vs 2K-L). ### Detection of secretory component associated with anti-Spike and anti-RBD antibodies in saliva from participants receiving COVID-19 mRNA vaccines The i.m. route induces an immune response in the axillary draining lymph node that is biased towards class switch to IgG rather than IgA. Thus, we were curious as to why we were able to detect an IgA anti-Spike/RBD response in the saliva, particularly after one dose of mRNA vaccine. We hypothesized that some IgA may be produced locally. To test this, we designed an ELISA to detect secretory chain associated with anti-Spike/RBD Ab. We determined that the secretory chain signal associated with anti-SARS-CoV-2 salivary Ab could be out-competed with recombinant secretory chain, and no anti-Spike/RBD secretory chain signal was detected in pre-pandemic colostrum, demonstrating assay specificity (Fig. S2). We then measured secretory component associated anti-SARS-CoV-2 Ab in the saliva of vaccinated LTCH participants who had received two doses of either BNT162b2 or mRNA-1273. We found that secretory component associated anti-Spike and anti-RBD Ab could be detected in 30% and 58% of participants, respectively, although the levels were significantly lower than what was observed in COVID-19 patients (Fig. 3A-B). The anti-SARS-CoV2 associated secretory chain signal was independent of prior SARS-CoV-2 exposure as we observed no significant difference in this signal comparing participants who were positive vs negative for serum anti-nucleocapsid protein Ab (Fig. 3C-D). Of note, if we divided the LTCH cohort into those participants who were positive versus negative for anti-Spike/RBD IgA, we observed that the secretory component signal was only detected in the IgA+ participants (Fig. 3E-F). Moreover, anti-Spike/RBD IgA and the level of secretory component positively correlated with each other at (Fig. S3). Combined with the finding that most participants did not produce IgM Ab to anti-Spike/RBD, we conclude that secretory component is associating with anti-Spike/RBD IgA (sIgA). Therefore, a local sIgA response to Spike/RBD is produced in response to mRNA vaccination in some participants. ![Fig 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/12/15/2021.08.01.21261297/F3.medium.gif) [Fig 3.](http://medrxiv.org/content/early/2021/12/15/2021.08.01.21261297/F3) Fig 3. Detection of secretory component associated with anti-Spike and anti-RBD antibodies in saliva from participants receiving COVID-19 mRNA vaccines. An ELISA-based method was used to detect secretory component associated with anti-Spike (**A**) and anti-RBD (**B**) antibodies in the saliva of 2-dose vaccinated subjects (n=93), as well as saliva taken from COVID-19 negative and positive patients (n=77 and 75, respectively). Subjects receiving 2 doses of BNT162b2 or mRNA1273 (**C**,**D**) were further grouped based on anti-NP antibody status, which is indicative of previous infection. Sub-setting of 2-dose vaccinated subjects into those that were considered above (n=34) or below (n=19) the positive cut off for salivary IgA analyzed for secretory component (**E**,**F**). Secretory component associated with anti-spike (**G**) and anti-RBD (**H**) antibodies in samples collected post-dose 1 and dose 2 from both MSB-1 (n=27) and MSB-2 (n=36). Solid black bars denote the median for each cohort, while the dotted black line denotes the positive cutoff, calculated as 2 standard deviations above the mean of a pool of negative control samples Mann-Whitney U test was used to calculate significance, with ns=not significant, **=p<0.01, \***|=p<0.001 and \**\*|\*=p<0.0001. ### Different decay kinetics of anti-Spike/RBD IgG versus IgA in saliva from participants receiving COVID-19 mRNA vaccines Since the LTCH workers were the earliest recipients of mRNA vaccines during the Canadian roll-out, we had the opportunity to examine the stability of anti-Spike/RBD IgG and IgA in the saliva of a subset of the LTCH cohort who are now reaching 6 months post-dose 2. We examined the level of salivary Ab only in those participants who had remained anti-Spike IgA positive at 2-weeks post-dose 2 (n=45, approximately 30% of participants). Using a paired analysis, we observed a significant decline in antigen-specific IgG and IgA levels in the saliva at this time point compared to 2-weeks post-dose 2, with the exception of anti-RBD IgA levels which trended downwards at 6-months post-dose 2 in participants who received mRNA-1273 (Fig. 4A-D). Anti-Spike Ab associated with the secretory component remained stable in both BNT162b2 and mRNA-1273 vaccinated subjects, although anti-RBD secretory component associated antibodies were significantly reduced at 6 months post-dose 2 (Fig. 4E-F). Therefore, most antibodies against SARS-CoV-2 in the saliva significantly decline over a 6-month period, with the exception of sIgA against Spike. ![Fig 4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/12/15/2021.08.01.21261297/F4.medium.gif) [Fig 4.](http://medrxiv.org/content/early/2021/12/15/2021.08.01.21261297/F4) Fig 4. Different decay kinetics of anti-Spike and anti-RBD IgG versus IgA in saliva from participants receiving COVID-19 mRNA vaccines. Saliva from LTCH participants was assessed for the presence of IgG (**A, B**), IgA (**C, D)** and sIgA (**E, F**) antibodies against Spike (A-C) or RBD (D-F). Mann-Whitney U test was used to calculate significance between groups. Only those participants who were positive for anti-Spike IgA at 2 weeks post-dose 2 (n=45) were analyzed. ns= not significant, *=p<0.05, \***|=p<0.001, and \**\*|\*=p<0.0001. ### Participants who experience a breakthrough infection have lower levels of vaccination-induced anti-Spike IgA Correlates of protection against SARS-CoV-2 breakthrough infection are ill-described and have only been examined for IgG10. Given that we have detected anti-Spike IgA in the serum and saliva of mRNA vaccinated participants, we examined whether IgA levels may be associated with protection against breakthrough infection. On 20th April 2021, an outbreak of P.1 lineage SARS-CoV-2 (gamma variant) was declared in a Toronto LTC home11. The residents were part of a separate serum antibody study following LTC residents that included sampling of blood, but not saliva, at 2-4 weeks post-dose 2 (received on February 2, 2021). Since anti-Spike IgA levels correlate with each other at 2 weeks post-dose 2 (Fig. S1), we also measured serum anti-Spike IgA at this time point as a proxy of the salivary IgA response. In the context of this isolated outbreak where n=5 residents were infected, we did not see a significant difference between exposed cases and exposed controls in terms of levels of anti-Spike/RBD IgG at 2-4 weeks post-dose 2 (Fig. 5A). We noted a trend of reduced anti-Spike IgA levels in cases versus controls that was significant by Mann Whitney analysis, but this significance did not survive the post-hoc Kruskal Wallis multiple test correction (Fig. 5B). ![Fig. 5.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/12/15/2021.08.01.21261297/F5.medium.gif) [Fig. 5.](http://medrxiv.org/content/early/2021/12/15/2021.08.01.21261297/F5) Fig. 5. Participants who experience a breakthrough infection have lower level of anti-Spike/RBD IgA at 2-4 weeks post-vaccination: Serum samples from vaccinated Toronto LTCH residents were taken at 2-4 weeks post-dose 2. Serum anti-Spike IgG (**A**) and IgA (**B**) levels were compared in participants who were subsequently exposed to P.1 gamma SARS-CoV-2 and either infected (case) or not infected (controls). In a separate cohort of double vaccinated health care workers from the Sheba Medical Center in Ramat Gan, Israel, serum samples were taken at 2-4 weeks post-dose 2. Serum anti-Spike IgG (**C**) and IgA (**D**) levels were compared in participants who experienced a breakthrough infection (cases) vs controls who did not. Solid black lines denote the median for each cohort. Kruskal-Wallis test with a correction for multiple comparisons was used to conduct statistical analysis between groups. NS=not significant, *=p<0.5, and **=p<0.01. Given the number of breakthrough infections in this isolated outbreak was small but nevertheless yielded a trend towards reduced anti-Spike IgA levels at 2 weeks post-dose 2 in exposed cases vs controls, we endeavored to test a replication cohort. We therefore measured anti-Spike serum IgA levels at approximately 2-4 weeks post-dose 2 in a larger cohort of health care workers (HCW) who had received 2 doses of BNT162b2 at the Sheba Medical Center in Ramat Gan, Israel10. Examining anti-Spike levels for each isotype, we found that IgG levels were modestly but not significantly lower in the serum of cases versus controls (Fig. 5C). In contrast, anti-Spike and anti-RBD IgA levels were both significantly lower in cases vs controls, and this held up to a multiple correction using a Kruskal Wallace Dunn’s multiple comparison test (Fig. 5D). In addition, we plotted the hazard ratio for participants according to their anti-Spike IgG and IgA levels and found that participants who had anti-Spike IgA levels greater than the cohort median were statistically less likely to experience a breakthrough infection during the observation period compared to those participants who were below the anti-Spike IgA cohort median (Fig. S4). Thus, high levels of anti-Spike serum IgA at 2-4 weeks post-dose 2 are associated with protection against subsequent breakthrough infection. ## Discussion In this study we observed robust IgG levels to Spike/RBD in the saliva of participants immunized with BNT162b2 or mRNA-1273 that correlate with the systemic IgG response, and these IgG are significantly diminished at 6 months post-dose 2. A different picture emerges, however, for salivary anti-Spike/RBD IgA. In most participants we observed a modest sIgA response to Spike/RBD in the saliva after a single dose of mRNA vaccine which is maintained in only approximately 30% of participants. Moreover, in these subjects the sIgA response to Spike is more resilient to decay. This preserved sIgA response may be very important for preventing breakthrough infections. Indeed, vaccinated subjects who subsequently experience a breakthrough infection have significantly lower levels of anti-Spike serum IgA at 2-4 weeks post-dose 2 and subjects with anti-Spike IgA levels are significantly less likely to acquire a breakthrough infection. Our findings of IgA in the saliva are consistent with recent reports showing that after two doses of BNT162b2 or mRNA-1273 anti-Spike and anti-RBD Ab could be detected in saliva12-14. We also distinguish between locally produced and systemically derived IgA using secretory component detection. It is unclear how anti-Spike/RBD sIgA is generated in the saliva following i.m. immunization. Of note, Spike protein can be detected in the plasma, increasing one to 5 days after mRNA-1273 vaccination using an ultra-sensitive detection technique15. Thus one possibility is that plasma-associated Spike antigen may reach the salivary glands (which are surrounded by capillaries16), provoking a local sIgA response. Another possibility is that a mucosal IgA response to mRNA vaccination takes place in the gut, and plasma cells generated at this location leave the gut (as we have shown before17), disseminating to other mucosal surfaces such as the oral cavity. Indeed, expression of the gut homing integrin α4β7 on circulating immune cells has been observed following administration of yellow fever18 and cholera toxin vaccines19 via the systemic route in humans. Recirculation between mucosal compartments could explain the sustained levels of anti-Spike sIgA in the saliva at 6 months post-boost which is not observed for the systemic IgG response. Animal models are ideal for obtaining further insights into mechanisms that explain how SARS-CoV-2 mRNA vaccines provoke antigen-specific sIgA production at mucosal surfaces. There are some limitations to our study: we did not have saliva available from the LTCH residents nor the Sheba HCW for confirmation that the association of high serum anti-Spike IgA levels with protection against breakthrough infection was also observed in a mucosal secretion. However, the levels of anti-Spike IgA in the saliva at 2 weeks post-dose 2 correlate with IgA levels in the serum (Fig. S1) suggesting that what we are detecting in the serum mirrors the oral compartment. Another limitation of our study is that we do not yet have 6-month samples examining the level of anti-Spike/RBD antibodies in the saliva of subjects who had a longer interval between dose 1 and dose 2. It will be of interest to determine if dose interval impacts salivary antibody decay rates. Lastly, it is unclear whether anti-Spike IgA antibodies in the saliva will further erode after 6 months post-dose 2. Dimeric secretory IgA has been shown to have potent neutralization activity against SARS-CoV-2 *in vitro*20. In this study we did not measure neutralizing activity of our saliva. Our samples have been tested by colleagues at Stanford University in a flow cytometry-based pseudovirus entry assay and it was determined that saliva from COVID-19 mRNA vaccinated subjects can modestly prevent viral infection of ACE2+ cells21. However, aside from direct neutralization, other IgA-dependent mechanisms may be relevant to protection against breakthrough infection. For example, IgA may serve to “enchain” microbes as it does in other mucosal sites such as the gut22, and even non-mucosal sites such as the brain meninges23. In summary, we provide evidence that anti-Spike sIgA are induced and maintained in the saliva of approximately 30% of mRNA vaccinated participants and that high anti-Spike IgA levels are associated with protection against subsequent breakthrough infection. Intranasal (i.n.) adenoviral-based vaccines that robustly induce anti-Spike/RBD IgA have been shown to prevent transmission in golden hamsters24. Moreover, a SARS-CoV-2 mRNA vaccine i.m. prime followed by an adenoviral i.n. boost provides superior protection against infection compared to i.m. only regimes, even against divergent variants such as B.1.1.351 (beta)25. An i.m. prime followed by i.n. boost strategy is worth considering for preventing person-to-person transmission of SARS-CoV-2 and for broad protection against emerging variants. ## Supporting information Supplemental Methods Figures and Tables [[supplements/261297_file07.pdf]](pending:yes) ## Data Availability The data will be available upon reasonable request. ## Acknowledgments Dr. Yves Durocher at the National Research Council of Canada (NRC) kindly donated reagents for the serum assays. Recombinant Vesicular Stomatitis Virus (rVSV)-eGFP-SARS-CoV-2-Spike was generously provided by Dr. Sean Whelan, and hACE2-mCherry expressing HEK293 cells was generously provided by Dr. Siyuan Ding, Washington University, St. Louis for neutralization assays performed by the Stanford scientists. We thank Elizabeth Yue, Antonio Estacio, Serena Loklam Chau, Ryan Law and Eric Yixiao Cao for LTCH sample processing and sustaining the infrastructure of sample processing in the Ostrowski lab. We thank Jeff Browning for critical reading of this manuscript. We would like to thank Florian Krammer for advice on secretory component detection by ELISA. Funding for this study was from a Foundation grant from the Canadian Institutes of Health Research (JG, Fund #15992), a COVID-19 Immunity Task force grant (SS, AM, MO, ACG and JG), an “Ontario Together” province of Ontario grant (JG and ACG), a Donation from the Royal Bank of Canada (RBC) Donation from the Krembil Foundation to the Sinai Health System Foundation. ## Footnotes * We have added a new Figure 5 and Supplemental Figure 4. * Received August 1, 2021. * Revision received December 15, 2021. * Accepted December 15, 2021. * © 2021, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## References 1. 1.Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT, Veesler D. Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell 2020;181(2):281–292 e6. DOI: 10.1016/j.cell.2020.02.058. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2020.02.058&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32155444&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F12%2F15%2F2021.08.01.21261297.atom) 2. 2.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(12):1177–1179. DOI: 10.1056/NEJMc2001737. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMc2001737&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32074444&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F12%2F15%2F2021.08.01.21261297.atom) 3. 3.Isho B, Abe KT, Zuo M, et al. Persistence of serum and saliva antibody responses to SARS-CoV-2 spike antigens in COVID-19 patients. Sci Immunol 2020;5(52). DOI: 10.1126/sciimmunol.abe5511. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImltbXVub2xvZ3kiO3M6NToicmVzaWQiO3M6MTM6IjUvNTIvZWFiZTU1MTEiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8xMi8xNS8yMDIxLjA4LjAxLjIxMjYxMjk3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 4. 4.Huang N, Perez P, Kato T, et al. SARS-CoV-2 infection of the oral cavity and saliva. Nat Med 2021;27(5):892–903. DOI: 10.1038/s41591-021-01296-8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-021-01296-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33767405&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F12%2F15%2F2021.08.01.21261297.atom) 5. 5.Stadtmueller BM, Huey-Tubman KE, Lopez CJ, Yang Z, Hubbell WL, Bjorkman PJ. The structure and dynamics of secretory component and its interactions with polymeric immunoglobulins. Elife 2016;5. DOI: 10.7554/eLife.10640. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.7554/eLife.10640&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26943617&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F12%2F15%2F2021.08.01.21261297.atom) 6. 6.Wang Z, Lorenzi JCC, Muecksch F, et al. Enhanced SARS-CoV-2 Neutralization by Secretory IgA in vitro. BioRxiv 2020;[https://www.biorxiv.org/content/10.1101/2020.09.09.288555v1](https://www.biorxiv.org/content/10.1101/2020.09.09.288555v1). 7. 7.Sterlin D, Mathian A, Miyara M, et al. IgA dominates the early neutralizing antibody response to SARS-CoV-2. MedRxiv 2020;[https://www.medrxiv.org/content/10.1101/2020.06.10.20126532v1](https://www.medrxiv.org/content/10.1101/2020.06.10.20126532v1). 8. 8.Fischinger S, Boudreau CM, Butler AL, Streeck H, Alter G. Sex differences in vaccine-induced humoral immunity. Semin Immunopathol 2019;41(2):239–249. DOI: 10.1007/s00281-018-0726-5. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00281-018-0726-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F12%2F15%2F2021.08.01.21261297.atom) 9. 9.Demonbreun AR, Sancilio A, Velez ME, et al. COVID-19 mRNA vaccination generates greater IgG levels in women compared to men. J Infect Dis 2021. DOI: 10.1093/infdis/jiab314. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/infdis/jiab314&link_type=DOI) 10. 10.Bergwerk M, Gonen T, Lustig Y, et al. Covid-19 Breakthrough Infections in Vaccinated Health Care Workers. N Engl J Med 2021;385(16):1474–1484. DOI: 10.1056/NEJMoa2109072. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2109072&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34320281&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F12%2F15%2F2021.08.01.21261297.atom) 11. 11.Williams C, Al-Bargash D, Macalintal C, et al. COVID-19 Outbreak Associated with a SARS-CoV-2 P.1 Lineage in a Long-Term Care Home after Implementation of a Vaccination Program - Ontario, April-May 2021. Clin Infect Dis 2021. DOI: 10.1093/cid/ciab617. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/cid/ciab617&link_type=DOI) 12. 12.Ketas TJ, Chaturbhuj D, Cruz-Portillo VM, et al. Antibody responses to SARS-CoV-2 mRNA vaccines are detectable in saliva. bioRxiv 2021. DOI: 10.1101/2021.03.11.434841. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMS4wMy4xMS40MzQ4NDF2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzEyLzE1LzIwMjEuMDguMDEuMjEyNjEyOTcuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 13. 13.Ketas TJ, Chaturbhuj D, Portillo VMC, et al. Antibody Responses to SARS-CoV-2 mRNA Vaccines Are Detectable in Saliva. Pathog Immun 2021;6(1):116–134. DOI: 10.20411/pai.v6i1.441. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.20411/pai.v6i1.441&link_type=DOI) 14. 14.Sano K, Bhavsar D, Singh G, et al. Efficient mucosal antibody response to SARS-CoV-2 vaccination is induced in previously infected individuals. medRxiv 2021:2021.12.06.21267352. DOI: 10.1101/2021.12.06.21267352. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMS4xMi4wNi4yMTI2NzM1MnYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMTIvMTUvMjAyMS4wOC4wMS4yMTI2MTI5Ny5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 15. 15.Ogata AF, Cheng CA, Desjardins M, et al. Circulating SARS-CoV-2 Vaccine Antigen Detected in the Plasma of mRNA-1273 Vaccine Recipients. Clin Infect Dis 2021. DOI: 10.1093/cid/ciab465. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/cid/ciab465&link_type=DOI) 16. 16.Zhang CZ, Cheng XQ, Li JY, et al. Saliva in the diagnosis of diseases. Int J Oral Sci 2016;8(3):133–7. DOI: 10.1038/ijos.2016.38. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ijos.2016.38&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F12%2F15%2F2021.08.01.21261297.atom) 17. 17.Rojas OL, Probstel AK, Porfilio EA, et al. Recirculating Intestinal IgA-Producing Cells Regulate Neuroinflammation via IL-10. Cell 2019;176(3):610–624 e18. DOI: 10.1016/j.cell.2018.11.035. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2018.11.035&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30612739&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F12%2F15%2F2021.08.01.21261297.atom) 18. 18.Masopust D, Choo D, Vezys V, et al. Dynamic T cell migration program provides resident memory within intestinal epithelium. J Exp Med 2010;207(3):553–64. DOI: 10.1084/jem.20090858. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamVtIjtzOjU6InJlc2lkIjtzOjk6IjIwNy8zLzU1MyI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzEyLzE1LzIwMjEuMDguMDEuMjEyNjEyOTcuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 19. 19.Quiding-Jarbrink M, Nordstrom I, Granstrom G, et al. Differential expression of tissue-specific adhesion molecules on human circulating antibody-forming cells after systemic, enteric, and nasal immunizations. A molecular basis for the compartmentalization of effector B cell responses. J Clin Invest 1997;99(6):1281–6. DOI: 10.1172/JCI119286. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI119286&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9077537&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F12%2F15%2F2021.08.01.21261297.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1997WQ61300021&link_type=ISI) 20. 20.Wang Z, Lorenzi JCC, Muecksch F, et al. Enhanced SARS-CoV-2 neutralization by dimeric IgA. Sci Transl Med 2021;13(577). DOI: 10.1126/scitranslmed.abf1555. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InNjaXRyYW5zbWVkIjtzOjU6InJlc2lkIjtzOjE1OiIxMy81NzcvZWFiZjE1NTUiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8xMi8xNS8yMDIxLjA4LjAxLjIxMjYxMjk3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 21. 21.G.R.N Salomon-Shulman, R.E.B.G., et al. Intramuscular SARS-CoV-2 vaccines elicit varying degrees of plasma and salivary antibody responses as compared to natural infection Medrxiv (In process) 2021. 22. 22.Bergwerk M, Gonen T, Lustig Y, et al. Covid-19 Breakthrough Infections in Vaccinated Health Care Workers. N Engl J Med 2021. DOI: 10.1056/NEJMoa2109072. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2109072&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34320281&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F12%2F15%2F2021.08.01.21261297.atom) 23. 23.Moor K, Diard M, Sellin ME, et al. High-avidity IgA protects the intestine by enchaining growing bacteria. Nature 2017;544(7651):498–502. DOI: 10.1038/nature22058. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature22058&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28405025&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F12%2F15%2F2021.08.01.21261297.atom) 24. 24.Langel SN, Johnson S, Martinez CI, et al. Oral and intranasal Ad5 SARS-CoV-2 vaccines decrease disease and viral transmission in a golden hamster model. bioRxiv 2021:2021.10.03.462919. DOI: 10.1101/2021.10.03.462919. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMS4xMC4wMy40NjI5MTl2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzEyLzE1LzIwMjEuMDguMDEuMjEyNjEyOTcuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 25. 25.Lapuente D, Fuchs J, Willar J, et al. Protective mucosal immunity against SARS-CoV-2 after heterologous systemic prime-mucosal boost immunization. Nat Commun 2021;12(1):6871. DOI: 10.1038/s41467-021-27063-4. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-021-27063-4&link_type=DOI)