SARS-CoV-2 infections elicit higher levels of original antigenic sin antibodies compared to SARS-CoV-2 mRNA vaccinations ======================================================================================================================== * Elizabeth M. Anderson * Theresa Eilola * Eileen Goodwin * Marcus J. Bolton * Sigrid Gouma * Rishi R. Goel * Mark M. Painter * Sokratis A. Apostolidis * Divij Mathew * Debora Dunbar * Danielle Fiore * Amanda Brock * JoEllen Weaver * John S. Millar * Stephanie DerOhannessian * The UPenn COVID Processing Unit * Allison R. Greenplate * Ian Frank * Daniel J. Rader * E. John Wherry * Scott E. Hensley ## SUMMARY Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccines elicit higher levels of antibodies compared to natural SARS-CoV-2 infections in most individuals; however, the specificities of antibodies elicited by vaccination versus infection remain incompletely understood. Here, we characterized the magnitude and specificity of SARS-CoV-2 spike-reactive antibodies from 10 acutely infected health care workers and 23 participants who received mRNA-based SARS-CoV-2 vaccines. We found that infection and primary mRNA vaccination elicited S1 and S2-reactive antibodies, while secondary vaccination boosted mostly S1 antibodies. Using magnetic bead-based absorption assays, we found that SARS-CoV-2 infections elicited a large proportion of original antigenic sin-like antibodies that bound efficiently to common seasonal human coronaviruses but poorly to SARS-CoV-2. In converse, vaccination only modestly boosted antibodies reactive to common seasonal human coronaviruses and these antibodies bound efficiently to SARS-CoV-2. Our data indicate that SARS-CoV-2 mRNA vaccinations elicit fundamentally different antibody responses compared to SARS-CoV-2 infections. ![Figure1](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/10/01/2021.09.30.21264363/F1.medium.gif) [Figure1](http://medrxiv.org/content/early/2021/10/01/2021.09.30.21264363/F1) **HIGHLIGHTS** * SARS-CoV-2 mRNA vaccines elicit higher levels of antibodies compared to SARS-CoV-2 infections * The first dose of an mRNA vaccine generates both S1 and S2 responses while the second dose boosts primarily S1-specific antibodies * SARS-CoV-2 infections, but not mRNA vaccinations, elicit high levels of antibodies that bind strongly to seasonal coronaviruses but weakly to SARS-CoV-2 KEYWORDS * SARS-CoV-2 * antibodies * mRNA vaccines * original antigenic sin * coronavirus ## BACKGROUND Since late 2019 severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread across the world causing a global pandemic1,2. This prompted the rapid development of several SARS-CoV-2 vaccines including two that use an mRNA-based platform (reviewed in3-5). The mRNA vaccines, Pfizer BNT162b2 and Moderna mRNA-1273, employ lipid nanoparticles that encase modified mRNA encoding the spike protein of SARS-CoV-26. SARS-CoV-2 mRNA vaccines have been found to be safe and effective at preventing severe COVID-19, hospitalizations, and death7,8. Recent studies demonstrate that SARS-CoV-2 mRNA vaccines elicit long-lived antibody responses that partially recognize and protect against antigenically distinct SARS-CoV-2 variants9-15. Some studies suggest that prior infections with common seasonal human coronaviruses (hCoVs) impact the severity of SARS-CoV-2 infections16,17. Most individuals are exposed to hCoVs early in childhood18-23 and then re-exposed to antigenically drifted forms of these viruses throughout life24-26. Common hCoVs include the HKU1 and OC43 betacoronaviruses (β-hCoVs) and 229E and NL63 alphacoronaviruses27-30. Studies from our group and others have shown that some individuals possessed antibodies that could bind to SARS-CoV-2 proteins prior to the COVID-19 pandemic18,31,32. SARS-CoV-2 is a β-hCoV, and antibodies reactive to the OC43 β-hCoV can be boosted upon SARS-CoV-2 infections16,18,32,33 and SARS-CoV-2 mRNA vaccinations34-36. It is unknown if the recall of β-hCoV antibodies upon SARS-CoV-2 infections impacts disease outcome. A recent study suggests that the recall of OC43 β-hCoV antibodies is associated with a compromised *de novo* SARS-CoV-2 response in individuals with fatal COVID-1937. The boosting of hCoV antibodies upon infection with the antigenically distinct SARS-CoV-2 is consistent with the doctrine of ‘original antigenic sin’, first proposed to describe influenza virus antibody responses by Thomas Francis in 196038. We recently developed new absorption assays to show that sequential heterosubtypic influenza virus infections elicit antibodies that paradoxically do not bind effectively to the boosting viral strain39. In the current report, we used a similar absorption technique to define the specificity of β-hCoV antibodies elicited by SARS-CoV-2 infections. We completed a series of studies to determine if these boosted β-hCoV antibodies could cross-react to SARS-CoV-2 and we compared antibodies elicited by SARS-CoV-2 infections and SARS-CoV-2 mRNA vaccinations. ## RESULTS ### SARS-CoV-2 infections and vaccinations elicit antibodies against the SARS-CoV-2 spike protein We obtained samples from individuals before and after acute SARS-CoV-2 infections (n=10) and pre-/post-two doses of a Pfizer BNT162b2 mRNA SARS-CoV-2 vaccine (n=23). Samples from SARS-CoV-2 infected individuals were obtained from a health care worker sero-monitoring study in which all infections were relatively mild16. All 10 individuals were seronegative in the beginning of this study and acquired a PCR-confirmed SARS-CoV-2 infection over the course of the study. Blood samples were collected from SARS-CoV-2 infected individuals 5-28 days (mean 17.9 days) after PCR-confirmed infections. Blood samples were collected from SARS-CoV-2 mRNA vaccinated individuals40 7-15 days post-primary immunization (mean 14.2 days), the day of or day before the booster immunization (mean 21.2 days post-primary), and 7-12 days post-booster immunization (mean 7.5 days post-boost). Individuals in our infection and vaccination studies had similar ages and both groups were predominantly female (**Supplemental Table 1**). View this table: [Supplemental Table 1](http://medrxiv.org/content/early/2021/10/01/2021.09.30.21264363/T2) Supplemental Table 1 Consistent with previous studies (reviewed in41), antibodies against the full-length spike (FL-S) of SARS-CoV-2 increased following infection (**Figure 1A**) and vaccination (**Figure 1B**). SARS-CoV-2 FL-S antibody levels were similar following infections and primary vaccinations, and antibody levels were significantly boosted following the second dose of vaccine (**Figure 1B-C**). The SARS-CoV-2 FL-S protein consists of two domains, the S1 domain which encompasses the receptor binding domain (RBD) essential for cell attachment and entry42-45 and the N terminal domain (NTD), and the S2 domain which shares more sequence homology with hCoVs28,33. Infection and primary vaccination elicited high levels of both S1 and S2 antibodies (**Figure 1A-B**), whereas secondary vaccinations boosted S1 antibodies more efficiently compared to S2 antibodies (**Figure 1B, D-E**). ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/10/01/2021.09.30.21264363/F2.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2021/10/01/2021.09.30.21264363/F2) Figure 1. Specificity of SARS-CoV-2 antibodies induced after SARS-CoV-2 infection versus vaccination. ELISAs were completed to quantify levels of serum antibodies binding to the SARS-CoV-2 full-length spike (FL-S) protein, the S1 domain (S1) of S, and the S2 domain (S2) of S after SARS-CoV-2 infection (**A)** and mRNA vaccination (**B**). Paired t-tests of log2 transformed antibody titers \**\*|\*p<0.0001, \***|p<0.001, **p<0.01, *p<0.05. We calculated fold-change in antibody titers before and after seroconversion and pre-/post-prime and boost doses of a SARS-CoV-2 mRNA vaccine (**C-E**). One way ANOVA of antibody fold change \**\*|\*p<0.0001, **p<0.01 *p<0.05. Horizontal lines indicate geometric mean and error bars represent standard deviation. ### SARS-CoV-2 infections and vaccinations elicit antibodies against a common human betacoronavirus We previously found that antibodies against the FL-S of the OC43 β-hCoV are boosted upon SARS-CoV-2 infection in hospitalized patients with severe disease18. Consistent with these findings, we found that antibodies reactive to the OC43 FL-S increased upon SARS-CoV-2 infections in health care workers with mild disease (**Figure 2A**). A recent study showed that antibody titers against β-hCoVs increase upon SARS-CoV-2 mRNA vaccination36. Although we found small increases in OC43 FL-S antibody titers following SARS-CoV-2 mRNA vaccinations (**Figure 2B**), the magnitude of OC43 FL-S antibody boosts were much lower following vaccinations compared to infections for most individuals (**Figure 2C**). Antibodies boosted by infections and vaccinations primarily targeted the S2 domain of the OC43 spike (**Figure 2A-B, 2D-E**), with infections boosting S2 responses more effectively compared to vaccinations (**Figure 2E**). Booster vaccinations did not affect levels of OC43 FL-S, S1, or S2 antibody levels (**Figure 2B-E**). ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/10/01/2021.09.30.21264363/F3.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2021/10/01/2021.09.30.21264363/F3) Figure 2. Antibodies to a related seasonal coronavirus are boosted upon SARS-CoV-2 infection and after vaccination to a lesser extent. ELISAs were completed to quantify levels of serum antibodies binding to the betacoronavirus, OC43 full-length spike (FL-S) protein, the S1 domain (S1) of S, and the S2 domain (S2) of S after SARS-CoV-2 infection (**A)** and mRNA vaccination **(B)**. Paired t-tests of log2 transformed antibody titers \**\*|\*p<0.0001, \***|p<0.001, **p<0.01, *p<0.05. We calculated fold-change in antibody titers before and after seroconversion and pre-/post- prime and boost doses of a SARS-CoV-2 mRNA vaccine (**C-E**). One way ANOVA of antibody fold change \**\*|\*p<0.0001 and \***|p=0.0001. Horizontal lines indicate geometric mean and error bars represent standard deviation. ### SARS-CoV-2 infections elicit higher levels of original antigenic sin antibodies compared to SARS-CoV-2 mRNA vaccinations We previously developed absorption-based assays to measure the level of cross-reactivity of antibodies elicited by influenza virus infections39. In that study, we sequentially infected ferrets with two antigenically distinct influenza virus strains and analyzed serum samples collected after each infection. We found that many antibodies elicited by secondary influenza virus infections paradoxically did not bind effectively to the secondary boosting influenza virus strain. We proposed that antigenically distinct influenza viruses engage memory B cells elicited by prior infections through multiple low affinity interactions with thousands of B cell receptors on memory B cells. Low affinity antibodies secreted in a soluble form through this recall response fail to bind to the antigenically distinct recall antigens because they require the level of multivalent binding that is provided on B cells. We developed a magnetic bead-based absorption assay to determine if OC43 FL-S antibodies elicited by SARS-CoV-2 infection and vaccination cross-react with SARS-CoV-2 FL-S. We incubated serum samples with beads coupled with either OC43 FL-S or SARS-CoV-2 FL-S and then we depleted bead-reactive antibodies using a magnetic column. Then, we quantified antibody levels in serum absorbed with antigen coupled beads to assess antibody cross-reactivity. SARS-CoV-2 FL-S-reactive antibodies elicited by infection were efficiently depleted with SARS-CoV-2 FL-S coupled beads but not OC43 FL-S coupled beads (**Figure 3A**). OC43 FL-S-reactive antibodies boosted by SARS-CoV-2 infection were efficiently depleted with OC43 FL-S coupled beads, but surprisingly these antibodies were not depleted with SARS-CoV-2 FL-S coupled beads (**Figure 3B**). These results are surprising since these OC43 FL-S-reactive antibodies were boosted upon SARS-CoV-2 infection. Vaccination-elicited antibodies had different cross-reactive properties compared to infection-elicited antibodies. Similar to antibodies elicited by infection, SARS-CoV-2 FL-S-reactive antibodies elicited by vaccination were efficiently depleted with SARS-CoV-2 FL-S coupled beads but not OC43 FL-S coupled beads (**Figure 3D**). In contrast to antibodies elicited by infection, OC43 FL-S-reactive antibodies boosted by SARS-CoV-2 vaccination were efficiently depleted with both OC43 FL-S and SARS-CoV-2 FL-S coupled beads (**Figure 3B**). As a control, we quantified levels of influenza virus hemagglutinin (HA) antibodies in these experiments and we found that HA-reactive antibodies were not depleted with SARS-CoV-2 FL-S or OC43 FL-S labeled beads (**Figure 3C, 3F**). Taken together, our data suggest that SARS-CoV-2 infection elicits OC43 FL-S-reactive antibodies that bind poorly to SARS-CoV-2 FL-S, while SARS-CoV-2 mRNA vaccinations elicits a relatively lower level of OC43 FL-S-reactive antibodies that efficiently cross-react with both OC43 FL-S and SARS-CoV-2 FL-S. ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/10/01/2021.09.30.21264363/F4.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2021/10/01/2021.09.30.21264363/F4) Figure 3. Seasonal coronavirus spike antibodies boosted by SARS-CoV-2 infection do not bind well to SARS-CoV-2 spike. Sera samples from 10 SARS-CoV-2 infected health care workers (**A-C**) and 10 SARS-CoV-2 mRNA vaccinated participants (**D-F**) were absorbed with SARS-CoV-2 full length (FL) S-coupled beads, OC43 Full length (FL) S-coupled beads, or mock treated beads prior to antibody quantification by ELISA. We determined reciprocal antibody titers in samples before and after infection and pre-/post- the first dose of an mRNA vaccine for SARS-CoV-2 FL-S (**A**,**D**), OC43 FL-S (**B**,**E**), and an unrelated antigen, influenza hemagglutinin H1 (**C, F**). Paired t-tests of log2 transformed antibody titers \**\*|\*p<0.0001, \***|p<0.001, **p<0.01. ## DISCUSSION Here we found that polyclonal antibodies elicited by SARS-CoV-2 mRNA vaccines are at a higher magnitude and have different specificities compared to those elicited by SARS-CoV-2 infections. We demonstrate that primary vaccinations elicit antibodies that bind to the S1 and S2 region of SARS-CoV-2 spike, and that S1-specific antibody responses are preferentially boosted after second vaccine doses. These data are consistent with recent reports that indicate that mRNA vaccinations elicit broader and more diverse antibody responses that are more effective at neutralizing SARS-CoV-2 variants compared to antibodies elicited by infection15,46-48,49. We found major differences in OC43 spike binding between antibodies elicited by SARS-CoV-2 infections versus mRNA vaccinations. SARS-CoV-2 infections elicited high levels of antibodies that bound to the S2 region of the OC43 spike protein; however, our absorption assays demonstrated that these antibodies bound poorly to the SARS-CoV-2 spike. Conversely, we found that SARS-CoV-2 mRNA vaccinations elicited lower levels of antibodies that reacted to the S2 region of the OC43 spike protein. Unlike antibodies elicited by infections, these vaccine-elicited antibodies were truly cross-reactive and bound efficiently to both SARS-CoV-2 and OC43 spike proteins. Further studies will be required to fully understand mechanisms that lead to different types of antibody responses elicited by SARS-CoV-2 infections versus vaccinations. It is possible that memory B cells elicited by prior β-hCoV infections are recalled by both SARS-CoV-2 infections and vaccinations, and that long-lived germinal centers elicited by mRNA vaccinations50 are required to allow for somatic hypermutations that promote the formation of cross-reactive S2 antibodies that bind efficiently to the spike proteins of both β-hCoVs and SARS-CoV-2. Consistent with this, a recent study found that S2-specific B cells with a memory phenotype are quickly recruited following primary immunization of humans51 Boosting of OC43 S2-reactive antibodies following SARS-CoV-2 infection is consistent with Thomas Francis’ doctrine of ‘original antigenic sin’38. Francis found that antibodies elicited by influenza vaccines often bound strongly to influenza virus strains that an individual was exposed to in childhood, although it is not apparent if these recalled influenza virus antibody responses typically occur at the expense of producing *de novo* antibodies (as we reviewed here52) The functional consequences of recalling low affinity S2-reactive antibodies following SARS-CoV-2 infections are unclear. Our previous studies found no correlation between OC43-reactive antibody induction and disease outcome following SARS-CoV-2 infection18 but a recent study suggested that the recall of OC43-reactive antibodies is associated with a compromised *de novo* SARS-CoV-2 response in individuals with fatal COVID-1937. Further studies are required to determine how the induction of different types of hCoV and SARS-CoV-2 antibodies affect disease outcome following SARS-CoV-2 infections. ## STAR METHODS ### KEY RESOURCES TABLE View this table: [Table1](http://medrxiv.org/content/early/2021/10/01/2021.09.30.21264363/T1) ## RESOURCES AVAILABILITY ### Lead Contact Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Scott E. Hensley (hensley{at}pennmedicine.upenn.edu). ### Materials Availability All unique reagents generated in this study will be available from the Lead Contact upon reasonable request. ### Data and Code Availability All raw data generated in this study will be deposited on Mendeley Data: ## EXPERIMENTAL MODEL AND SUBJECT DETAILS ### Samples from Human Subjects The infection cohort described in this report consists of health care workers within the University of Pennsylvania Healthcare System who were recruited into a SARS-CoV-2 sero-monitoring study that included biweekly blood draws as previously described16. A nasal pharyngeal (NP) swab was collected from all health care workers who tested positive for SARS-CoV-2 IgG and or IgM antibodies or who were experiencing COVID-like symptoms during the study period. NP swabs were PCR tested for the presence of SARS-CoV-2 viral RNA. Seroconverted health care workers with PCR-confirmed SARS-CoV-2 infection (n=10 adults >18 years old) were included in this analysis. The vaccination cohort described in this report consists of participants (n=23 adults >18 years old) who enrolled in a study at the University of Pennsylvania that included blood draws before and after two vaccination doses with an mRNA-based COVID-19 vaccine as previously described40. Whole blood was collected from participants who provided proof of vaccination with Pfizer (BNT162b2) mRNA vaccines. Samples were collected at 4 timepoints: 1-2 weeks before vaccination (baseline) (visit 1; V1), 1-2 weeks post-primary immunization (visit 2; V2), the day of or day before booster immunization (visit 3; V3), and 1-2 weeks post-booster immunization (visit 4; V4). Only participants without prior SARS-CoV-2 exposure were included in this report. Plasma and PBMCs were isolated from whole blood for downstream assays. All sera and plasma samples were heat-inactivated in a 56°C water bath for 1 hour prior to serological testing. All samples were collected after obtaining informed consent and studies were approved by the University of Pennsylvania Institutional Review Board. ### Cell lines 293F cells were from Thermo fisher (Thermo Fisher cat. R79007). 293T cells were from ATCC (ATCC cat. CRL-3216, RRID:CVCL_0063). All cell lines were cultured using manufacturer’s guidelines and used as described in Method Details below. ## METHOD DETAILS ### Proteins for serological studies SARS-CoV-2 full length spike (FL-S) protein was purified by Ni-NTA resin from 293F cells transfected with a plasmid that encodes the FL-S (A gift from Florian Krammer, Icahn School of Medicine at Mt. Sinai, New York City NY)53. S1 and S2 subunits of the SARS-CoV-2 spike were purchased from Acro Biosystems (ACROBiosystems, Newark, DE; cat. S1N-C52H3, and S2N-C52H5, respectively) and reconstituted in 200 μL Dulbecco’s phosphate buffered saline (DPBS) to a final concentration of 500 μg/mL. OC43 FL-S was also purchased (Sino Biological, Wayne PA; cat. 40588-V08B) and reconstituted in DPBS. OC43 subunit proteins were purified in our laboratory as previously described18. Briefly, mammalian expression plasmids encoding the S1 (amino acids 15-760) or S2 (amino acids 766-1305) domains of the OC43 spike protein were cloned with an N-terminal OC43 S signal peptide, and a C-terminus encoding a Factor Xa cleavage site, a trimerization domain from T4 fibritin (Foldon), a site-specific biotinylation sequence (AviTag), and a hexa-histidine purification tag. 293F cells were transfected with S1 or S2 encoding plasmids and proteins were purified from cell culture supernatant 6 days later with Ni-NTA resin (Qiagen, Hilden, Germany). Proteins were concentrated and buffer exchanged into PBS with Amicon centrifugal filters (Millipore, Burlington, MA) prior to quantification on a spectrophotometer (NanoDrop, Thermo Fisher Scientific, Waltham, MA). ### ELISAs Antibodies reactive to SARS-CoV-2 and OC43 antigens were quantified by enzyme-linked immunosorbent assays (ELISA) as previously described18,54. Absorbed sera samples were also tested for the presence of influenza virus H1 HA antibodies. In brief, ELISA plates (Thermo Fisher Scientific, Waltham, MA: cat. 14-245-153) were coated overnight at 4°C with either 2 μg/mL SARS-CoV-2 or influenza HA antigens, 1.5 μg/mL OC43 antigens, or Dulbecco’s phosphate buffered saline (DPBS) to control for background antibody binding. Sera was heat-inactivated in a 56°C water bath for 1 hour prior to serial dilutions starting at 1:50 in dilution buffer. ELISA plates were blocked for 1 hour before 50 μL of diluted sera was added and plates were incubated for 2 hours on an orbital shaker. ELISA plates were washed 3 times with 1x PBS supplemented with 2% Tween (PBS-T) before the addition of goat anti-human IgG conjugated to horseradish peroxidase secondary antibody at a 1:5000 dilution (Jackson ImmunoResearch Laboratories, West Grove, PA: cat. 109-036-098). ELISA plates were developed with TMB substrate, and the reactions were stopped after 5 minutes by the addition of 250 mM hydrochloric acid prior to reading on a SpectraMax 190 microplate reader (Molecular Devices, San Jose, CA). Serum antibody titers were obtained from a standard curve of either serially diluted monoclonal antibody (CR3022 for SARS-CoV-2 or CR9114 for influenza virus HA starting at 0.5μg/mL) or serially diluted pooled serum (for OC43 ELISAs). Standard curves were included on every plate to control for plate-to-plate variation. Antibody titers for each sample were measured in at least two technical replicates performed on separate days. ### Carboxyl magnetic bead absorptions SARS-CoV-2 FL-S or OC43 FL-S antigens were coupled to carboxyl magnetic beads (RayBiotech, Peachtree Corners, GA; cat. 801-114-2) at a concentration of 35 μg antigen/100 μL magnetic beads. Mock beads were prepared by the addition of DPBS in place of antigen. Briefly, 175 μg of diluted antigen or PBS (for mock) was added to 500 μL of magnetic beads and the mixture was incubated for 2 hours at 4°C with constant mixing. The unbound fractions were removed using a magnetic stand and conjugated beads were quenched by the addition of 300 μL 50 mM Tris, pH 7.4 prior to a 15-minute incubation at room temperature with constant mixing. Quenching buffer was removed using a magnetic stand and the conjugated beads were washed 4 times with 300 μL wash buffer (DPBS supplemented with 0.1% BSA and 0.05% Tween-20). After the final wash, beads were resuspended in 300 μL wash buffer and were stored at 4°C prior to use in serum absorption assays. Sera samples were absorbed with beads coupled to SARS-CoV-2 FL-S, OC43 FL-S, and mock beads. Sera samples were diluted in PBS to a final dilution of 1:25. Next, 20 μL of antigen coupled-magnetic beads or mock-treated beads were added to 100 μL of diluted sera and the mixtures were incubated for 1 hour at room temperature on a plate mixer at 800rpm. Fractions containing the unabsorbed antibodies were removed using a 96-well plate magnetic stand. Unabsorbed fractions were diluted in buffer (DPBS supplemented with 1% milk and 0.1% Tween-20) prior to running in ELISA. ## Data Availability All data are included in the manuscript. ## QUANTIFICATION AND STATISTICAL ANALYSIS Statistical analyses were performed using Prism version 8 (GraphPad Software, San Diego CA). Reciprocal serum dilution antibody titers were log2 transformed for statistical analysis. ELISA antibody titers below the limit of detection (LOD) were set to a reciprocal titer equal to half the LOD. Log2 transformed antibody titers were compared with paired and unpaired t-tests, and one-way ANOVAs with Tukey’s multiple comparisons. Statistical significance was defined as a p-value <0.05. ## DECLARATION OF INTERESTS E.J.W. has consulting agreements with and/or is on the scientific advisory board for Merck, Elstar, Janssen, Related Sciences, Synthekine and Surface Oncology. E.J.W. is a founder of Surface Oncology and Arsenal Biosciences. E.J.W. is an inventor on a patent (U.S. patent number 10,370,446) submitted by Emory University that covers the use of PD-1 blockade to treat infections and cancer. S.E.H. has received consultancy fee from Sanofi Pasteur, Lumen, Novavax, and Merck for work unrelated to this report. ## AUTHOR CONTRIBUTIONS Serological Assays, E.M.A., T.E., E.C.G, and M.J.B., Data Analyses, E.M.A. Cohort Studies and Sample Processing, S.G., R.R.G, M.M.P., S.A.A., D.M., D.D., D.F., A.B., J.W., J.M., S.D., the UPenn COVID Processing Unit, and A.R.G. Manuscript Writing, E.M.A., and S.E.H.; Supervision, I.F., D.J.R, E.J.W., and S.E.H.; Funding Acquisition, I.F., D.J.R, E.J.W., and S.E.H. ## ACKNOWLEDGEMENTS This project has been funded in part with Federal funds from the National Institute of Allergy and Infectious Diseases, National Institutes of Health, Department of Health and Human Services, under Contract No. 75N93021C00015. This work was supported in part by institutional funds from the University of Pennsylvania and NIH U19AI082630 (S.E.H. and E.J.W.). E.M.A. was supported by the NIH Training in Virology T32 Program (T32-AI-007324). We thank J. Lurie, J. Embiid, J. Harris, and D. Blitzer for philanthropic support. We thank all members of the Penn COVID-19 Sample Processing Unit. We would like to thank David Anderson for assistance with the graphical abstract. The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official views of the NIH. ## Footnotes * 10 The UPenn COVID Processing Unit is a unit of individuals from diverse laboratories at the University of Pennsylvania who volunteered time and effort to enable study of COVID-19 patients during the pandemic. Members are listed in the acknowledgement section. * Received September 30, 2021. * Revision received September 30, 2021. * Accepted October 1, 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. Carvalho, T., Krammer, F. & Iwasaki, A. The first 12 months of COVID-19: a timeline of immunological insights. Nat Rev Immunol 21, 245–256, doi:10.1038/s41577-021-00522-1 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41577-021-00522-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33723416&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 2. Zhou, P. et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 579, 270–273, doi:10.1038/s41586-020-2012-7 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-020-2012-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32015507&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 3. Excler, J. L., Saville, M., Berkley, S. & Kim, J. H. Vaccine development for emerging infectious diseases. Nat Med 27, 591–600, doi:10.1038/s41591-021-01301-0 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-021-01301-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 4. Gebre, M. S. et al. Novel approaches for vaccine development. Cell 184, 1589–1603, doi:10.1016/j.cell.2021.02.030 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2021.02.030&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33740454&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 5. Golob, J. L., Lugogo, N., Lauring, A. S. & Lok, A. S. SARS-CoV-2 vaccines: a triumph of science and collaboration. JCI Insight, doi:10.1172/jci.insight.149187 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/jci.insight.149187&link_type=DOI) 6. Topol, E. J. Messenger RNA vaccines against SARS-CoV-2. Cell 184, 1401, doi:10.1016/j.cell.2020.12.039 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2020.12.039&link_type=DOI) 7. Baden, L. R. et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N Engl J Med 384, 403–416, doi:10.1056/NEJMoa2035389 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2035389&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33378609&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 8. Polack, F. P. et al. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N Engl J Med 383, 2603–2615, doi:10.1056/NEJMoa2034577 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2034577&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33301246&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 9. Collier, D. A. et al. Sensitivity of SARS-CoV-2 B.1.1.7 to mRNA vaccine-elicited antibodies. Nature 593, 136–141, doi:10.1038/s41586-021-03412-7 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-021-03412-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33706364&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 10. Doria-Rose, N. et al. Antibody Persistence through 6 Months after the Second Dose of mRNA-1273 Vaccine for Covid-19. N Engl J Med 384, 2259–2261, doi:10.1056/NEJMc2103916 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMc2103916&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33822494&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 11. Liu, Y. et al. Neutralizing Activity of BNT162b2-Elicited Serum. N Engl J Med 384, 1466–1468, doi:10.1056/NEJMc2102017 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMc2102017&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33596352&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 12. Wang, Z. et al. mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants. Nature 592, 616–622, doi:10.1038/s41586-021-03324-6 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-021-03324-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33567448&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 13. Widge, A. T. et al. Durability of Responses after SARS-CoV-2 mRNA-1273 Vaccination. N Engl J Med 384, 80–82, doi:10.1056/NEJMc2032195 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMc2032195&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33270381&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 14. Wu, K. et al. Serum Neutralizing Activity Elicited by mRNA-1273 Vaccine. N Engl J Med 384, 1468–1470, doi:10.1056/NEJMc2102179 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMc2102179&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33596346&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 15. Wang, Z. et al. Naturally enhanced neutralizing breadth against SARS-CoV-2 one year after infection. Nature 595, 426–431, doi:10.1038/s41586-021-03696-9 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-021-03696-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34126625&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 16. Gouma, S. et al. Health care worker sero-monitoring reveals complex relationships between common coronavirus antibodies and COVID-19 symptom duration. JCI Insight, doi:10.1172/jci.insight.150449 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/jci.insight.150449&link_type=DOI) 17. Sagar, M. et al. Recent endemic coronavirus infection is associated with less-severe COVID-19. J Clin Invest 131, doi:10.1172/JCI143380 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI143380&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32997649&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 18. Anderson, E. M. et al. Seasonal human coronavirus antibodies are boosted upon SARS-CoV-2 infection but not associated with protection. Cell 184, 1858–1864 e1810, doi:10.1016/j.cell.2021.02.010 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2021.02.010&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33631096&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 19. Dijkman, R. et al. The dominance of human coronavirus OC43 and NL63 infections in infants. J Clin Virol 53, 135–139, doi:10.1016/j.jcv.2011.11.011 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jcv.2011.11.011&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22188723&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 20. Dyrdak, R., Hodcroft, E. B., Wahlund, M., Neher, R. A. & Albert, J. Interactions between seasonal human coronaviruses and implications for the SARS-CoV-2 pandemic: A retrospective study in Stockholm, Sweden, 2009-2020. J Clin Virol 136, 104754, doi:10.1016/j.jcv.2021.104754 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jcv.2021.104754&link_type=DOI) 21. Gaunt, E. R., Hardie, A., Claas, E. C., Simmonds, P. & Templeton, K. E. Epidemiology and clinical presentations of the four human coronaviruses 229E, HKU1, NL63, and OC43 detected over 3 years using a novel multiplex real-time PCR method. J Clin Microbiol 48, 2940–2947, doi:10.1128/JCM.00636-10 (2010). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNtIjtzOjU6InJlc2lkIjtzOjk6IjQ4LzgvMjk0MCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzEwLzAxLzIwMjEuMDkuMzAuMjEyNjQzNjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 22. Huang, A. T. et al. A systematic review of antibody mediated immunity to coronaviruses: kinetics, correlates of protection, and association with severity. Nat Commun 11, 4704, doi:10.1038/s41467-020-18450-4 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-020-18450-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32943637&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 23. Killerby, M. E. et al. Human coronavirus circulation in the United States 2014-2017. J Clin Virol 101, 52–56, doi:10.1016/j.jcv.2018.01.019 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jcv.2018.01.019&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29427907&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 24. Edridge, A. W. D. et al. Seasonal coronavirus protective immunity is short-lasting. Nat Med 26, 1691–1693, doi:10.1038/s41591-020-1083-1 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-020-1083-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32929268&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 25. Eguia, R. T. et al. A human coronavirus evolves antigenically to escape antibody immunity. PLoS Pathog 17, e1009453, doi:10.1371/journal.ppat.1009453 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.ppat.1009453&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33831132&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 26. Kistler, K. E. & Bedford, T. Evidence for adaptive evolution in the receptor-binding domain of seasonal coronaviruses OC43 and 229e. Elife 10, doi:10.7554/eLife.64509 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.7554/eLife.64509&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33463525&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 27. Jaimes, J. A., Andre, N. M., Chappie, J. S., Millet, J. K. & Whittaker, G. R. Phylogenetic Analysis and Structural Modeling of SARS-CoV-2 Spike Protein Reveals an Evolutionary Distinct and Proteolytically Sensitive Activation Loop. J Mol Biol 432, 3309–3325, doi:10.1016/j.jmb.2020.04.009 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jmb.2020.04.009&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32320687&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 28. Okba, N. M. A. et al. Severe Acute Respiratory Syndrome Coronavirus 2-Specific Antibody Responses in Coronavirus Disease Patients. Emerg Infect Dis 26, 1478–1488, doi:10.3201/eid2607.200841 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3201/eid2607.200841&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32267220&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 29. Andersen, K. G., Rambaut, A., Lipkin, W. I., Holmes, E. C. & Garry, R. F. The proximal origin of SARS-CoV-2. Nat Med 26, 450–452, doi:10.1038/s41591-020-0820-9 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-020-0820-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32284615&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 30. Boni, M. F. et al. Evolutionary origins of the SARS-CoV-2 sarbecovirus lineage responsible for the COVID-19 pandemic. Nat Microbiol 5, 1408–1417, doi:10.1038/s41564-020-0771-4 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41564-020-0771-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32724171&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 31. Ng, K. W. et al. Preexisting and de novo humoral immunity to SARS-CoV-2 in humans. Science 370, 1339–1343, doi:10.1126/science.abe1107 (2020). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzNzAvNjUyMi8xMzM5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMTAvMDEvMjAyMS4wOS4zMC4yMTI2NDM2My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 32. Song, G. et al. Cross-reactive serum and memory B-cell responses to spike protein in SARS-CoV-2 and endemic coronavirus infection. Nat Commun 12, 2938, doi:10.1038/s41467-021-23074-3 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-021-23074-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34011939&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 33. Nguyen-Contant, P. et al. S Protein-Reactive IgG and Memory B Cell Production after Human SARS-CoV-2 Infection Includes Broad Reactivity to the S2 Subunit. mBio 11, doi:10.1128/mBio.01991-20 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1128/mBio.01991-20&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32978311&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 34. Jackson, L. A. et al. An mRNA Vaccine against SARS-CoV-2 - Preliminary Report. N Engl J Med 383, 1920–1931, doi:10.1056/NEJMoa2022483 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2022483&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32663912&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 35. Roltgen, K. et al. mRNA vaccination compared to infection elicits an IgG-predominant response with greater SARS-CoV-2 specificity and similar decrease in variant spike recognition. medRxiv, doi:10.1101/2021.04.05.21254952 (2021). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMS4wNC4wNS4yMTI1NDk1MnYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMTAvMDEvMjAyMS4wOS4zMC4yMTI2NDM2My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 36. Amanat, F. et al. SARS-CoV-2 mRNA vaccination induces functionally diverse antibodies to NTD, RBD, and S2. Cell 184, 3936–3948 e3910, doi:10.1016/j.cell.2021.06.005 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2021.06.005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34192529&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 37. McNaughton, A. L. et al. Fatal COVID-19 outcomes are associated with an antibody response targeting epitopes shared with endemic coronaviruses. medRxiv, 2021.2005.2004.21256571, doi:10.1101/2021.05.04.21256571 (2021). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMS4wNS4wNC4yMTI1NjU3MXYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMTAvMDEvMjAyMS4wOS4zMC4yMTI2NDM2My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 38. Francis, T. On the Doctrine of Original Antigenic Sin. Proceedings of the American Philosophical Society 104, 572–578 (1960). 39. Arevalo, C. P. et al. Original antigenic sin priming of influenza virus hemagglutinin stalk antibodies. Proc Natl Acad Sci U S A 117, 17221–17227, doi:10.1073/pnas.1920321117 (2020). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTE3LzI5LzE3MjIxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMTAvMDEvMjAyMS4wOS4zMC4yMTI2NDM2My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 40. Goel, R. R. et al. Distinct antibody and memory B cell responses in SARS-CoV-2 naive and recovered individuals following mRNA vaccination. Sci Immunol 6, doi:10.1126/sciimmunol.abi6950 (2021). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImltbXVub2xvZ3kiO3M6NToicmVzaWQiO3M6MTM6IjYvNTgvZWFiaTY5NTAiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8xMC8wMS8yMDIxLjA5LjMwLjIxMjY0MzYzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 41. Lombardi, A. et al. Mini Review Immunological Consequences of Immunization With COVID-19 mRNA Vaccines: Preliminary Results. Front Immunol 12, 657711, doi:10.3389/fimmu.2021.657711 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fimmu.2021.657711&link_type=DOI) 42. Barnes, C. O. et al. Structures of Human Antibodies Bound to SARS-CoV-2 Spike Reveal Common Epitopes and Recurrent Features of Antibodies. Cell 182, 828–842 e816, doi:10.1016/j.cell.2020.06.025 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2020.06.025&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32577645&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 43. Letko, M., Marzi, A. & Munster, V. Functional assessment of cell entry and receptor usage for SARS-CoV-2 and other lineage B betacoronaviruses. Nat Microbiol 5, 562–569, doi:10.1038/s41564-020-0688-y (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41564-020-0688-y&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32094589&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 44. Walls, A. C. et al. Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell 181, 281–292 e286, doi:10.1016/j.cell.2020.02.058 (2020). [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%2F10%2F01%2F2021.09.30.21264363.atom) 45. Wrapp, D. et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 367, 1260–1263, doi:10.1126/science.abb2507 (2020). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzNjcvNjQ4My8xMjYwIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMTAvMDEvMjAyMS4wOS4zMC4yMTI2NDM2My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 46. Greaney, A. J. et al. Antibodies elicited by mRNA-1273 vaccination bind more broadly to the receptor binding domain than do those from SARS-CoV-2 infection. Sci Transl Med 13, doi:10.1126/scitranslmed.abi9915 (2021). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InNjaXRyYW5zbWVkIjtzOjU6InJlc2lkIjtzOjE1OiIxMy82MDAvZWFiaTk5MTUiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8xMC8wMS8yMDIxLjA5LjMwLjIxMjY0MzYzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 47. Starr, T. N. et al. SARS-CoV-2 RBD antibodies that maximize breadth and resistance to escape. Nature, doi:10.1038/s41586-021-03807-6 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-021-03807-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=438709&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 48. Stamatatos, L. et al. mRNA vaccination boosts cross-variant neutralizing antibodies elicited by SARS-CoV-2 infection. Science, doi:10.1126/science.abg9175 (2021). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzNzIvNjU0OS8xNDEzIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMTAvMDEvMjAyMS4wOS4zMC4yMTI2NDM2My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 49. Planas, D. et al. Reduced sensitivity of SARS-CoV-2 variant Delta to antibody neutralization. Nature, doi:10.1038/s41586-021-03777-9 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-021-03777-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34237773&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 50. Turner, J. S. et al. SARS-CoV-2 mRNA vaccines induce persistent human germinal centre responses. Nature 596, 109–113, doi:10.1038/s41586-021-03738-2 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-021-03738-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34182569&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 51. Brewer, R. C., Ramadoss, N. S., Lahey, L. J., Robinson, W. H. & Lanz, T. V. BNT162b2 Vaccine Induces Divergent B cell responses to SARS-CoV-2 S1 and S2. medRxiv, 2021.2007.2020.21260822, doi:10.1101/2021.07.20.21260822 (2021). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMS4wNy4yMC4yMTI2MDgyMnYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMTAvMDEvMjAyMS4wOS4zMC4yMTI2NDM2My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 52. Cobey, S. & Hensley, S. E. Immune history and influenza virus susceptibility. Curr Opin Virol 22, 105–111, doi:10.1016/j.coviro.2016.12.004 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.coviro.2016.12.004&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28088686&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 53. Amanat, F. et al. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nat Med, doi:10.1038/s41591-020-0913-5 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-020-0913-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32398876&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F10%2F01%2F2021.09.30.21264363.atom) 54. Flannery, D. D. et al. SARS-CoV-2 seroprevalence among parturient women in Philadelphia. Sci Immunol 5, doi:10.1126/sciimmunol.abd5709 (2020). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImltbXVub2xvZ3kiO3M6NToicmVzaWQiO3M6MTM6IjUvNDkvZWFiZDU3MDkiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8xMC8wMS8yMDIxLjA5LjMwLjIxMjY0MzYzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==)