Hybrid adaptive immunity to SARS-CoV-2 protects against breakthrough infection after COVID-19 vaccination in ALSPAC participants ================================================================================================================================ * Holly E. Baum * Marianna Santopaolo * Ore Francis * Emily Milowdowski * Katrina Entwistle * Elizabeth Oliver * Benjamin Hitchings * Divya Diamond * Amy C. Thomas * Ruth E. Mitchell * Milla Kibble * Kapil Gupta * Natalie Di Bartolo * Paul Klenerman * Anthony Brown * Begonia Morales-Aza * Jennifer Oliver * Imre Berger * Ash M. Toye * Adam Finn * Anu Goenka * Andrew D. Davidson * Sue Ring * Lynn Molloy * Melanie Lewcock * Kate Northstone * Firona Roth * Nicholas J. Timpson * Linda Wooldridge * Alice Halliday * Laura Rivino ## SUMMARY Immunological memory to vaccination and viral infection involves coordinated action of B and T-cells, thus integrated analysis of these two components is critical for understanding their contributions to protection against breakthrough infections (BI). We investigated cellular and humoral immune responses to SARS-CoV-2 infection and/or COVID-19 vaccination in participants from the Avon Longitudinal Study of Parents and Children (ALSPAC). The magnitude of antibody and T-cell responses following the second vaccine dose was associated with protection against BI in participants with a history of SARS-CoV-2 infection (cases), but not in infection-naïve controls. Youden’s index thresholds for protection against BI were calculated for all immune measures. Anti-Spike IgG (>666.4 BAU/mL) and anti-nucleocapsid N pan Ig (>0.1332 BAU/mL) thresholds combined were 100% sensitive and 83% specific for cases without BI over 8-months follow-up. Collectively these results point to the superior protective effect of hybrid immunity and have implications for the design of next-generation COVID-19 vaccines. KEYWORDS * ALSPAC * SARS-CoV-2 * COVID-19 * adaptive immunity * infection * vaccination * hybrid immunity * breakthrough infection ## INTRODUCTION The COVID-19 pandemic caused substantive medical and socioeconomic burdens that continue to affect the global population 1–4. Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) presents as a broad spectrum of clinical manifestations from asymptomatic or mild infections, through to severe illness, and associated mortality 5,6. The rapid deployment of COVID-19 vaccines proved instrumental in reducing SARS-CoV-2 infections and subsequent hospitalisations and deaths, however vaccine effectiveness wanes over time 7–9. Breakthrough infections (BIs) are well documented and are influenced not only by waning immunity, but also the evolution of novel viral variants 10–13. A broad repertoire of antibody and cellular responses are elicited by SARS-CoV-2 infection 14–16 and COVID-19 vaccination 17–19. Evidence for Spike-specific IgG and neutralizing antibody titres as correlates of protection (COPs) against SARS-CoV-2 infection and severe disease have been demonstrated in COVID-19 vaccine trials 20,21, and studies of BIs 12,13. Additionally, both systemic and mucosal IgA have been implicated in protection against infection 22–24. SARS-CoV-2-specific T-cells are more durable than antibodies 25, and may therefore be important for protection longer-term, and against novel variants which can evade antibody-mediated immunity 26,27. Furthermore, cross-reactive T-cells can enhance infection- and vaccine-mediated responses 28 and induce abortive infections in highly exposed individuals29. Anti-viral immune memory responses are determined by the coordinated action of antibodies and T-cells targeting the virus, and an integrated analysis of these two components is therefore critical for understanding their contributions to protection. In this study we investigate the cellular and humoral responses to SARS-CoV-2 after infection and/or vaccination in participants from the Avon Longitudinal Study of Parents and Children (ALSPAC). We further address the role of antibodies and T-cells in providing protection against BI in participants with hybrid immunity, induced by a combination of SARS-CoV-2 infection and COVID-19 vaccination, as compared to those with vaccine-induced immunity alone. ## METHODS ### ALSPAC The Avon Longitudinal Study of Parents and Children (ALSPAC) is a birth cohort study 30–33. Pregnant women resident in Avon, UK with expected dates of delivery between 1st April 1991 and 31st December 1992 were invited to take part. A total of 14,541 pregnancies were initially enrolled, with 13,988 children who were alive at 1 year of age. ALSPAC now comprises three generations: the original pregnant women with the biological fathers and other carers/partners (G0), the cohort of index children (G1), and the offspring of the index children (G2). This has generated a wealth of biological, genetic and phenotypic data spanning the lifetime of these individuals. Including additional participants recruited in the interim period, the total sample size for analyses using any data collected after the age of seven is 15,447 pregnancies, resulting in 15,658 foetuses. Of these 14,901 children were alive at 1 year of age. 12,113 G0 partners have been in contact with the study, of which 3,807 are currently enrolled. Please note that the study website contains details of all the data that are available through a fully searchable data dictionary and variable search tool 34. From the beginning of the COVID-19 pandemic, ALSPAC sought to utilise their unique expertise and infrastructure to contribute to SARS-CoV-2 research efforts through collection of biological samples and questionnaire data from their cohort of well characterised participants. ### Study design In October 2020, ALSPAC undertook 5200 serological SARS-CoV-2 lateral flow tests (LFTs) on G0 and G1 cohort participants. Participants with evidence of a previous SARS-CoV-2 infection based on a positive IgG result on this LFT, and/or a positive SARS-CoV-2 PCR result from linked UK Health Security Agency (UKHSA) data, were invited to take part in the study (n=124). Two control groups with negative serological results, and no documented positive PCR test, were recruited alongside these participants. The first control group (n=93) were also age, sex, and symptom (anosmia) matched to those with a history of SARS-CoV-2 infection. Participants in the second control group (n=103) were selected on the basis of not having reported anosmia. Full details of the recruitment methodology are presented in (ref. paper 1). Participants attended 3 clinics in December 2020 (clinic 1), March 2021 (clinic 2) and June 2021 (clinic 3) where they provided venous blood and saliva samples. Health and lifestyle information was gathered through online questionnaires. The characteristics of the cohort at clinic 1 (pre-vaccination) are described in detail in (ref. paper 1). For the purposes of the analyses presented in this study, the two control groups have been combined as no significant differences in baseline antibody or T-cell measures were detected between the two original groups (ref. paper 1). ### Ethics Ethical approval for the study was obtained from the ALSPAC Ethics and Law Committee and the Local Research Ethics Committees (NHS REC 20/HRA/4854). Consent for biological samples was collected in accordance with the Human Tissue Act (2004). Informed consent for the use of data collected via questionnaires and clinics was obtained from participants following the recommendations of the ALSPAC Ethics and Law Committee at the time. At age 18, study children were sent ‘fair processing’ materials describing ALSPAC’s intended use of their health and administrative records and were given clear means to consent or object via a written form. Data were not extracted for participants who objected, or who were not sent fair processing materials. Ethical approval for the study was obtained from the ALSPAC Law and Ethics committee and local research ethics committees (NHS Haydock REC: 10/H1010/70). ### Sample collection and processing Peripheral blood mononuclear cells (PBMCs) were obtained from up to 3 x 10 ml EDTA tubes per participant using standard density gradient separation techniques. Briefly, samples were kept at room temperature for up to 3 hours after drawing blood. Blood was diluted 1:1 with Phosphate Buffered Saline (PBS) containing 1% Fetal calf serum (FCS). Diluted blood was separated using a Ficoll gradient, centrifuging at 1000g for 10 mins at room temperature. PBMCs were washed in PBS/1% FCS, centrifuging at 700g for 10 mins at room temperature, and washed again with PBS/1% FCS, centrifuging at 400g for 10 mins at room temperature. Cell pellets were resuspended in freezing mix (90% FCS/10% DMSO) at a concentration of 13-15x106 cells per ml. Cells were frozen overnight in an alcohol bath to control freezing rate. PBMCs were then transferred to liquid nitrogen cryotanks for long-term storage. Serum tubes were left to clot, centrifuged at 1,500g for 10mins at 18-25°C, aliquoted, and stored at -80 °C. Participants provided neat saliva directly into a sterile collection tube. Particulate matter was removed by centrifugation at 13,000g for 10 minutes. Samples were heat-inactivated at 56°C for 30 minutes prior to ELISA analysis. ### ELISAs SARS-CoV-2-specific anti-Spike (S) IgA and IgG in serum and saliva were measured by ELISA. Samples were run in duplicate at a single optimised dilution and reported in BAU/mL following calibration of an internal standard to the WHO/NIBSC reference control. Serum anti-nucleocapsid (N) and anti-S pan Ig were utilised as screening ELISAs to report a positive/negative result for previous SARS-CoV-2 infection with data presented as normalised optical density (OD) measurement relative to an internal control. Thresholds for positivity were calculated on a large sample of PCR-confirmed and pre-pandemic samples. Full details of these methods are published for serum 35, and saliva 36, respectively. ### Synthetic peptides Peptides used for PBMC stimulations in the ELISpot assays are listed in Supplementary Table S1. 15-mer peptides overlapping by 10 amino acids and spanning the sequences of the SARS-CoV-2 S, N, Membrane (M) and Envelope (E) protein were purchased from Mimotopes (Australia). The purity of the peptides was >80% and their composition was confirmed by mass spectrometry analysis. 15-18mer peptides overlapping by 10 amino acids and spanning sequences of SARS-CoV-2 NSP1-16, ORF3, ORF6, ORF7 and ORF 8 were also purchased from Mimotopes (Australia). The purity of the peptides was ∼70%. Combination of peptides and identification of peptide pools are described in Table S1. For PBMC stimulation in ELISpot assays, pools of a maximum of 127 peptides were added per well as follows: Spike was divided into 2 pools (S1; 126 peptides and S2; 127 peptides); for smaller peptide regions, peptides were combined in the same peptide pools as follows: NSP1 and NSP2 (NSP1+2), NSP5 and NSP6 (NSP5+6), NSP15 and NSP16 (NSP15+16). All other peptides were pooled into one mixture and tested in individual ELISpot wells. ### Enzyme-linked immune absorbent spot assay (ELISpot) Human IFN-γ ELISpot assays were performed using a Human IFN-γ ELISpot BASIC kit (Mabtech). MSIP4W10 PVDF plates (Millipore) were coated with capture antibody (mAb-1-D1K; 15 μg/mL) and incubated overnight at 4 °C in carbonate bicarbonate buffer (Sigma Aldrich). Cryopreserved PBMC were thawed then rested at 37 °C/5% CO2 for 5-6 hours. Coated plates were washed 5 times in sterile PBS and blocked for 1-2 hours using R10 medium (0.2 µm filtered RPMI 1640 medium supplemented with 10 % FBS, 2 mM glutamine, penicillin ((100 units/ml) and streptomycin (100 μg/ml)). 4 x 105 PBMCs were added to the plate with or without peptide pools (as indicated) in a total assay volume of 100 µl in R10. PBMC incubated with R10 medium alone were used as negative (unstimulated) controls and were performed in duplicate. Peptide pools spanning S1, S2, M, N E, ORF1 (NSP1+2, NSP3A, NSP3B, NSP3C, NSP4, NSP5+6, NSP7-11, NSP12A, NSP12B, NSP13, NSP14, NSP15+16), ORF3, ORF6, ORF7 and ORF8 were used at a final concentration of 2 µg/ml. PBMC stimulated with anti-CD3 antibody (Mabtech, Mab CD3-2; final concentration 0.1% v/v) were used as a positive control for each participant using 1-4 x 105 PBMC per well. Positive control and peptide stimulated wells were performed in singlet. Plates were incubated for 16-18 hours at 37 °C/5% CO2 then developed as per manufacturer’s instructions. Developed plates were protected from light and air-dried for 48 hours before image acquisition using a CTL ImmunoSpot S6 Ultra-V Analyzer. Spot forming units (SFU) were calculated using the ImmunoSpot S6 Ultra-V Analyzer Basic Count function after image acquisition using optimised counting parameters that were applied across all participants. Spot counts were enumerated for each peptide pool by subtraction of average background (calculated from duplicate unstimulated wells). Counts were expressed as SFU per million (106) PBMC after multiplication by 2.5 following background subtraction. Negative values after background subtraction were adjusted to zero 29. Participants were excluded if the spot count in unstimulated wells exceeded 95 SFU per million PBMC or if no spots were observed in the positive control wells. Where spot formation was too dense to accurately enumerate using standardised counting parameters (TNTC; too numerous to count), affected wells were excluded unless contemporaneous assessment of IFN-γ production by flow cytometry confirmed antigen specific response. In these cases, TNTC values were given the raw value equivalent to the largest spot count accurately counted for a peptide pool (320 SFU per well). ### Intracellular Cytokine Staining (ICS) PBMCs were thawed and rested overnight in AIMV 2% FCS then incubated with or without peptide pools from SARS-CoV-2 S1/S2, M, N, NSP3B (all 1 μg/ml), or with PMA/ionomycin (PMA 10 ng/ml, ionomycin 100 ng/ml, Sigma-Aldrich) for 5 hr at 37°C in the presence of brefeldin A (BD, 5 μg/ml). To assess degranulation, anti CD107a-FITC antibody was added to the cells at the beginning of the stimulation. Cells were stained with a viability dye Zombie Aqua (BioLegend) for 10 min at room temperature and then with antibodies targeting surface markers (20 min 4°C, diluted in PBS 1% BSA [Sigma-Aldrich]). Cells were fixed overnight in eBioscience Foxp3/Transcription factor fixation/permeabilization buffer (Invitrogen), and intracellular staining was performed for detection intracellular cytokines, including IFN-γ, TNF-α, IL-2 and MIP1β (30 min 4°C). Four samples were excluded from further CD4+ analysis due technical issues. Data were acquired on a BD LSR Fortessa X20 and analysed using FlowJo software v10.8.1. A complete list of antibodies is included in Table S2. ICS was performed on samples from 69 individuals in the case group, selected based on T-cell response detectable by IFN-γ ELISpot for at least one for SARS-CoV-2 peptide pools tested and for whom we had additional cryopreserved PBMC vials available at baseline. ### Pseudoneutralisation Serum neutralisation was expected to positively correlate with levels of anti-Spike antibody binding results. Accordingly, all samples from participants in the case groups corresponding to anti-Spike pan-Ig ELISA results above a normalised threshold of 0.5 were included. Additionally, 8 samples immediately below this threshold (0.46-0.494) and a single sample far below this threshold (0.22) were included to reflect neutralisation kinetics of low anti-Spike antibody binding. For pseudovirus assays, Wuhan-Spike-harbouring pseudovirus (VSV-S-FLuc) was generated and used to assess serum antibody neutralisation of VSV-S-FLuc entry into Vero ACE2 TMPRSS2 (VAT) cells as described in *Halliday et al., 2022*. Briefly, serum dilutions starting at 1/40 followed by 8 2.5-fold titrations were plated in triplicate in 96-well plates, alongside three wells each of 1/25 dilutions of known neutralising and non-neutralising controls (corresponding to 16,000 RLU of luminescence when mixed with VSV-S-FLuc). WT Wuhan spike pseudotyped VSV was added to each well and incubated for an hour. Well-mixtures were added to black, microscopy 96-well plates, seeded with 10,000 Vero ACE2 TMPRSS2 (VAT) cells per well. Luminescence measurements were taken 16 hours after infection, using the ONE-Glo Luciferase Assay System (Promega no. E6120). ### Data analysis Study data were collected and managed using REDCap electronic data capture tools hosted at the University of Bristol. REDCap (Research Electronic Data Capture) is a secure, web-based software platform designed to support data capture for research studies 37. Statistical analyses were performed using R Studio (v4.3.0), and GraphPad Prism (version 10.01). Unpaired comparisons across multiple groups were done with the Kruskal-Wallis test with Dunn’s post-test for multiple comparisons. Pairwise correlations were assessed with Spearman’s rank-order correlation (rs). Correlation coefficients were interpreted as: weak (rs=0.20-0.39), moderate (rs=0.40-0·59), strong (rs=0.60-0.79), or very strong (rs=0.80-1.00). The following adjusted P value thresholds were used for data visualisation: P≤0.05 (*), P≤0.01 (**), P≤0.001 (\***|), P≤0.0001 (\**\*|\*). ## RESULTS ### Study design and recruitment This study was open to ALSPAC participants of the G0 (48-70 years) and G1 (29-30 years) generations. 377 participants enrolled, and attended clinics in December 2020, March 2021, and June 2021 (Figure S1). This samples drawn from the ALSPAC cohort was 59.2% female, 97.8% white, and 58.5% G1 generation (Table S3). Participants were selected from a cohort of 4819 individuals who undertook SARS-CoV-2 antibody lateral flow testing (LFT) in October 2020 (Figure S2). LFT anti-S IgG positivity rates in the G0 (48-70 years) and G1 (29-30 years) generations were 3.0% and 5.6% in respectively. These results were used as an initial screen to identify participants with and without a likely history of SARS-CoV-2 infection for inclusion in this study (ref paper 1). To validate the LFT results, serum samples from clinic 1 were screened for SARS-CoV-2-specific anti-S and anti-N antibodies using in-house ELISAs 35. Participants were defined as ‘cases’ if they had a previous PCR-confirmed SARS-CoV-2 infection and/or were positive on either ELISA (Figure 1A). The remaining seronegative individuals with no history of COVID-19 are herein referred to as ‘controls’. Clinic 1 samples were taken prior to participants being vaccinated as part of the UK COVID-19 vaccination programme. Of those who provided information on their vaccination status 64.0% had received at least one dose by clinic 2, rising to 97.0% by clinic 3 (Figure S3). Data was therefore analysed relative to the number of COVID-19 vaccinations received, rather than chronologically by clinic, as this was deemed to be the dominant variable influencing immune responses (Figure S4). ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/06/15/2024.06.14.24308948/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2024/06/15/2024.06.14.24308948/F1) Figure 1: Antibody and T-cell responses to COVID-19 vaccination in SARS-CoV-2 naïve and previously infected individuals. (A) Participants were classified as cases based on a previous PCR-confirmed SARS-CoV-2 infection and/or positivity on the serum anti-Spike (S) and or anti-Nucleocapsid (N) pan Ig screening ELISAs. (B) Serum pseudoneutralising antibody titres. (C, D) Anti-S IgG and IgA in serum and saliva, measured by ELISA. Correlation coefficients calculated using Spearman’s rank (rs). (E) Magnitude of SARS-CoV-2 specific T-cell responses against S (pool-1 and 2) N, M and NSP3B peptide pools measured via ELISpot assay. Red bars indicate median responses. Unpaired comparisons were performed using Kruskal-Wallis test with Dunn’s correction for multiple comparisons. Within each of the case and control groups, responses were compared between 0 and 1, and 1 and 2 vaccine doses. Responses between groups were compared after each dose. Statistics are only displayed for comparisons where P ≤0.05 (*), ≤0.01 (**), ≤0.001 (\***|), ≤0.0001 (\**\*|\*). ### Antibody responses to COVID-19 vaccination At baseline, anti-S and anti-N pan Ig levels were higher in cases compared to controls (P≤0.0001; Figure 1A). A vaccine-induced increase in anti-S pan Ig was observed in both groups. Following the second dose, all participants had anti-S pan Ig levels above the assay positivity threshold. However, the case group retained their baseline advantage with higher median titres. Rates of new SARS-CoV-2 infections between clinics 1-3 were low, with only four individuals in the control group reporting a positive PCR test. Consistent with this observation, a small increase in median anti-N pan Ig was observed after the first dose (P=0.0471), which plateaued after dose 2 (Figure 1A). This was predominantly driven by ≥2-fold increases in anti-N pan Ig levels in a small minority of participants (3/95; Figure S5). Anti-N pan Ig levels in the case group remained stable throughout. Anti-S IgG in serum and saliva showed similar kinetics to serum anti-S pan Ig responses (Figure 1C). Post-vaccination anti-S IgG levels in the two samples were very strongly correlated after the first (rs=0.858, P≤0.0001) and second (rs=0.809, P≤0.0001) doses. Serum pseudoneutralising antibody titres in the case group increased in response to the first vaccination, then remained stable following the second dose (Figure 1B). A strong positive correlation between anti-S IgG and pseudoneutralising antibody titre was observed following each dose (rs=0.784, P≤0.0001 / rs=0.656, P≤0.0001; Figure S6). Pre-vaccination anti-S IgA in serum and saliva was higher in cases than controls (Figure 1D). Serum anti-S IgA increased in both groups after the first dose, whilst median salivary anti-S IgA levels were not boosted by vaccination. There was therefore only a weak correlation between the two measures post-vaccination (rs=0.355, P≤0.0001 / rs=0.246, P=0.0045). Recipients of mRNA vaccines had higher median serum anti-S IgG and IgA levels than recipients of the adenoviral vector based ChAdOx1 (Figure S7). ### T-cell responses to COVID-19 vaccination T-cell responses targeting SARS-CoV-2 S1/S2, N, M, NSP3B, non-structural proteins (NSP3-16) and accessory proteins (ORF3, ORF7 and ORF8) were measured by IFN-γ ELISpot (Figure S7A-C). Baseline S-specific IFN-γ responses were lower in controls compared to cases (P≤0.0001; Figure 1E). In both groups, S-specific T-cells increased after the first vaccination and then remained stable. T-cell responses to N, M and NSP3B were not boosted by vaccination in the case group. In contrast, increased T-cell responses to M (P≤0.0001), N (P=0.0175), and NSP3B (P=0.0091) were observed in control participants after dose 2 compared to baseline (Figure 1E). T-cell responses to NSP3-16, ORF3, ORF7 and ORF8, remained stable following vaccination (Figure S8A/B). ### Magnitude and quality of T-cell responses to SARS-CoV-2 infection The magnitude and features of the CD4+ and CD8+ T-cell response targeting S, N, M and NS3PB proteins were analysed in cases, prior to vaccination. Production of IFN-γ, TNF-α, MIP1β, IL-2 and degranulation (CD107a) by SARS-CoV-2-specific CD4+ and CD8+ T-cells was measured by ICS. A representative gating strategy is shown in Figure S9. Responses were comprised of monofunctional and polyfunctional T-cells producing 1 or >1 effector functions respectively, with monofunctional T-cells dominating both the CD4+ and CD8+ responses (Figure 2A/B). Monofunctional SARS-CoV-2-specific CD4+ T-cells produced mainly TNF-α (S1=59.03%, S2=28.45%, M=25.7%, N=41.35%, NSP3B=27.35%) and IFN-γ (S1=23.59%, S2=16.75%, M=35.39%, N=34.55%, NSP3B=48.34%). The frequencies of CD4+ T-cells producing each cytokine were comparable across proteins (Figure S10A). In contrast, CD107a was expressed at higher levels by CD4+ T-cells targeting M (34.26%), compared to S1 (5.25%), S2 (5.23%), and N proteins (9.42%) (Figure S10A). Polyfunctional CD4+ T-cells were mainly IFN-γ+/TNF-α+, IL-2+/TNF-α+ and IFN-γ+/IL-2+/TNFα+ (Figure 2A). ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/06/15/2024.06.14.24308948/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2024/06/15/2024.06.14.24308948/F2) Figure 2: SARS-COV-2-specific CD4+ and CD8+ T-cell responses in previously infected individuals. The magnitude and quality of CD4+ (A) and CD8+ (B) T-cell responses targeting Spike, M, N and NSP3B proteins measured by intracellular cytokine staining. Significance was determined using a Kruskal-Wallis test with Dunn’s correction for multiple comparisons. Similarly to CD4+ T cells, cytokine production by CD8+ T-cells targeting the different SARS-CoV-2 proteins was comparable while CD107a was expressed by a larger proportion of CD8+ T-cells specific for M compared to other proteins (M=31.7%, S1=1.63%, S2=4.91%, N=3.52%, NSP3B=4.27%) (Figure S12B). Polyfunctional CD8+ T-cells were mainly double or triple-cytokine producing, including IFN-γ+/TNF-α+, CD107a+/TNF-α+, MIP1β+/TNF-α+, MIP1β+/IFN-γ+ and MIP1β+/IFN-γ+/TNF-α+. NSP3B-specific CD8+ T-cells displayed higher polyfunctionality (14.8%) compared to those targeting S (S1=10.4%, S2=5.29%) or (N=5.33%) (Figure 2B; Figure S12A/B). ### Association between post-vaccination immune responses and protection against SARS-CoV-2 BI We next investigated the relationship between the magnitude of antibody and T-cell responses after the second vaccination, and protection against SARS-CoV-2 BI. The case and control groups were subdivided based on self-reported questionnaire data detailing whether the participant had a PCR/LFT-confirmed SARS-CoV-2 infection in the 8-months following the sample collection period (July 2021 - March 2022; Figure S1). For this analysis, the case group was updated to include two individuals who had PCR-confirmed SARS-CoV-2 infections between clinics 1-3. BI rates were higher in the control group (42.4%) compared to the cases (30.8%). Of the individuals who reported BIs, the median time since receiving a second vaccination, and the proportion who received a third vaccination prior to infection, were comparable between cases and controls (Table S4). Self-reported BIs were consistent with increased rates of LFT anti-N IgG positivity detected in the wider ALSPAC cohort in the period from May 2021 (G1=14.6%; G0=9.8%) to May 2022 (G1=67.1%; G0=50.3%; Figure S2). In participants without history of COVID-19, there was no strong evidence of differences between those who went on to experience a BI, compared to those who didn’t, with respect to the magnitude of immune variables measured (Figure S12A; Figure 3A). Conversely, previously infected participants showed evidence of clustering by subsequent infection status (Figure 3B). Higher levels of Spike-specific IgG in serum and saliva, pseudoneutralising antibody, and anti-S IgA in serum as well as S1-specific T-cell responses were observed in those with no reported BI (Figure S12B). Similarly, high anti-N Pan Ig titres were associated with a decreased likelihood of re-infection. The lack of vaccine-induced salivary IgA in this cohort was reflected in the poor performance of this measure as a discriminator of future infection susceptibility. ![Figure 3:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/06/15/2024.06.14.24308948/F3.medium.gif) [Figure 3:](http://medrxiv.org/content/early/2024/06/15/2024.06.14.24308948/F3) Figure 3: Post-vaccination antibody and T-cell levels correlate with a reduced susceptibility for reported SARS-CoV-2 breakthrough infection in previously infected participants. Correlation of post-second vaccination immune responses in participants who self-reported a SARS-CoV-2 infection in the subsequent 8-months (breakthrough infection; BI) and those who didn’t (no breakthrough infection). (A) SARS-CoV-2-naïve individuals with (n=18) or without (n=25) a BI. (B) Previously SARS-CoV-2-infected individuals with (n=18) or without (n=25) a BI. See supplementary Figure 13 for derivation of the thresholds. To compare the effectiveness of individual markers for discriminating between cases who did or didn’t go on to be re-infected, ROC curves were plotted, and thresholds calculated using the Youden’s Index method to balance sensitivity and specificity (Figure S13). Anti-S IgG ≥666.4 BAU/mL in serum (SP=87.5%), or 0.547 BAU/mL in saliva (SP=75.0%), provided 94.44% sensitivity for identifying participants who didn’t report BIs. Specificity was improved to 100.0% by combining serum S-specific IgG and N-specific pan Ig thresholds (SE=83.3%, SP=100%). Combining S-specific serum IgG with IgA in serum or saliva, or with T-cell responses, did not improve discrimination. Pseudoneutralising antibody ≥6165 units provided a highly specific threshold (SP=100%) for identification of protected individuals, albeit with lower sensitivity (SE=61.1%) than the S-specific IgG threshold (SE=94.4%). The magnitude of T-cell responses to the S1 pool (SE=72.2%, SP=87.5%) provided a more specific and sensitive marker of infection susceptibility than those to the S2 (SE=66.67%, SP=75.0%) or N (SE=38.9%, SP 87.5%). Combining thresholds for T-cell S1 and N did not improve performance compared to S1 alone. Conversely, combining T-cell S1 with N-specific pan Ig offered improved specificity with no loss of sensitivity (SP=100%, SE=72.2%). ### Association between ICS responses to pre-vaccination SARS-CoV-2 infection and reported BI To determine whether specific features of the T-cell response to SARS-CoV-2 infections were also a factor in determining risk of BI, pre-vaccination ICS responses were compared in cases with and without a reported BI. We chose to include monofunctional CD4+ and CD8+ T cell responses in these analyses, as these were the predominant responses observed in these individuals. There was no meaningful difference between the magnitude of S, N or NS3PB-specific T-cell responses in the two groups (Figure 4A/B, Figure S11C/D). In contrast, participants who didn’t report BIs displayed higher magnitudes of SARS-CoV-2 M-specific CD4+ and CD8+ T-cells, and these were skewed respectively towards production of IFN-γ+ and CD107a (CD4+ 40.0% versus 23.1%; CD8+ 42.4% versus 4.9%; Figure 4C/D). M-specific CD4+ and CD8+ T-cells displayed higher MIP-1β production in individuals who developed BI. However, including these ICS measures in the analysis did not improve the predictive ability of thresholds generated using antibody and/or T-cell data alone. ![Figure 4:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/06/15/2024.06.14.24308948/F4.medium.gif) [Figure 4:](http://medrxiv.org/content/early/2024/06/15/2024.06.14.24308948/F4) Figure 4: Pre-vaccination SARS-CoV-2-specific T cell responses in cases with and without reported breakthrough infection measured by ICS assay. (A, B) Magnitude of single cytokine/CD107a-producing (monofunctional) SARS-CoV-2-specific CD4+ (A; total n= 47) and CD8+ (B; total n=51) T-cells specific for the indicated SARS-CoV-2 proteins among cases with and without reported breakthrough infection at baseline (pre-vaccination). Data in the A-B are shown as mean ± SD. Statistics were calculated by t-test (Mann-Whitney). (C, D) Pie charts depicting the proportions of single cytokine/CD107a-producing CD4+ (C; n=40) and CD8+ (D; n=43) T-cells within the respective M-specific T cell population. ## DISCUSSION Humoral and cellular immune responses to COVID-19 vaccination were measured in ALSPAC participants with and without a history or serological evidence of SARS-CoV-2 infection. In those with hybrid immunity, we demonstrate a correlation between the magnitude of responses following the second vaccination and protection against BI. The combination of serum S IgG (>666.4 BAU/mL) and N pan Ig (>0.1332 BAU/mL) thresholds identified those who didn’t report BIs with 100% sensitivity and 83% specificity. Our results suggest that hybrid immunity to SARS-CoV-2 remains effective in protection from reinfection at >15 months post infection and >8 months post second-dose vaccination. The reduced association in control participants alludes to the importance of the greater quality and breath of immune responses elicited by SARS-CoV-2 infection compared to vaccination alone. COVID-19 vaccination elicited robust serum anti-S IgG and IgA production in all participants, with the case group retaining their baseline immunological advantage as observed in other studies 38. mRNA vaccination resulted in higher S-specific IgG and IgA compared to the viral vector ChAdOx1, as previously reported 39. However, neither vaccine type induced salivary IgA, even in participants with a history of COVID-19 where vaccination has been suggested to boost infection-primed responses 22,40,41. Reducing or eliminating transmission with vaccines capable of eliciting mucosal immunity therefore remains a priority, and a number are currently in development 42. Consistent with recent papers, COVID-19 vaccines induced S-specific T-cell responses in all participants 43–45. Upon vaccination, the frequency of IFN-γ producing T-cells specific for NSP1-2 increased amongst cases, while in controls there was an increase in the frequency of T-cell responses to M, N, and NSP3B. As these proteins are not present in the vaccine formulations, these data suggest potential bystander T-cell activation as a result of vaccination. Antigen-independent bystander activation of T-cells which is likely driven by cytokines, has been widely reported during viral infection 46,47, however its occurrence in the context of vaccination remains unclear48–52. We cannot exclude that the observed increase in T-cell responses to non-spike SARS-CoV-2 proteins is due to asymptomatic, abortive SARS-CoV-2 infection, which was shown to boost T-cell responses in the absence of seroconversion 29. Long-term protection against SARS-CoV-2 BI relies upon the durability of vaccine-induced responses. Antibodies in particular have been shown to wane rapidly in the 6-months following the primary and subsequent vaccine doses 53–55. This declining immunity, particularly in the context of highly transmissible SARS-CoV-2 variants with reduced neutralising sensitivity, increases BI rates 8,10,56. Anti-S IgG and neutralising antibody levels have been shown to negatively correlate with the risk of SARS-CoV-2 infection and severe disease 12,21,57–59. In control participants, we observed no evidence of difference in the magnitude of responses between those who did and didn’t experience BIs, and protective thresholds could therefore not be defined. However, anti-S IgG ≥666.4 BAU/mL were associated with reduced incidence of BI in those with a history of SARS-CoV-2 infection. This is consistent with the large Israeli COVID-19 family study (ICoFS) which proposed S-specific IgG >500 BAU/mL, and neutralising antibody titres of >1024, as thresholds of protection against SARS-CoV-2 Delta infection 60. These levels were associated with an 11% probability of infection, and 1% probability of moderate disease. ICoFS baseline measurements were taken shortly before infection and therefore their lower threshold value is expected relative to our use of post-vaccination dose 2 titres that were recorded months earlier. Serum anti-S IgA also associated with reduced BIs in our cohort, and alongside mucosal IgA has previously been shown to correlate with protection against infection independently of IgG 22. BIs experienced shortly after primary vaccination correlated with lower vaccine-induced IgA 22, and in those who experienced infections serum IgA levels inversely correlated with symptom duration 61. Increased S-specific T-cells also correlated with reduced BI in our cohort, consistent with previous reports for both CD4+ and CD8+ responses 62–64. Furthermore, the combination of high neutralizing antibodies and S-specific IFN-γ responses is associated with protection against BI 65,66. There is now mounting evidence that hybrid immunity affords more robust and sustained protection against BI compared to vaccine-induced immunity alone 53,67–70. Consistent with this, BI rates in our study were lower in cases (30.8%) compared to controls (42.4%). Both humoral and cellular immune responses have been reported to be qualitatively superior in those who experience SARS-CoV-2 infection prior to vaccination 69,70, and infection-derived immunity may be particularly important in the context of BIs with novel variants 23,71. It is only SARS-CoV-2 infection that induces a significant and durable systemic IgA response 55, and COVID-19 vaccines perform poorly at eliciting mucosal IgA 41. Additionally, the increased magnitude of neutralising responses in hybrid immunity enhances the breadth of neutralising activity against divergent variants of concern 72,73. Vaccine-induced Fc-receptor binding antibodies are also important for the control and clearance of infection and have also been shown to be more abundant in those with a history of COVID-19, with this difference persisting post-vaccination 69. Several papers have reported that functional and phenotypic properties of SARS-CoV-2 adaptive immunity differ in response to SARS-CoV-2 infection and COVID-19 vaccination 74,75. Vaccinated individuals displayed a skewed T helper (Th)1 Ag-specific T-cell polarization and higher activated memory B-cells expressing IgG compared to those of patients who recovered from COVID-19 76. In this study we have only assessed the magnitude of previously infected individuals at baseline (pre-vaccination) specifically, we compared the magnitude of post-infection SARS-CoV-2-specific T-cell responses between individuals who did and didn’t go on to experience BIs. We show trends towards higher magnitudes of SARS-CoV2-specific CD8+ T-cells targeting S2, N, M and NSP3B in individuals who do not develop BI, with differences being significantly higher only for M, suggesting a protective role for SARS-CoV2 CD8+ T-cells. Similarly, M-specific CD4+ T-cells are present at higher magnitudes in individuals who do not develop BI compared to those that do. In addition, the former group displays M-specific CD4+ and CD8+ T-cells producing mainly IFN-γ and CD107a, which suggests a protective role of cytotoxic Th1-Tc1 T-cells in COVID-19. These differences were not detected using IFN-γ ELISPOT, which shows higher magnitudes of S1-specific T-cells in individuals who do not develop BI, but similar levels of T-cells targeting the other SARS-CoV-2 proteins. These data highlight how more granular analyses of T-cell responses are needed to define T-cell features associated with protection in COVID-19. This will be critical if we aim to incorporate T-cell responses into our definitions of the correlates and determinants of protective immunity to SARS - CoV-2 and potentially other acute viral infections. In addition to changing the quality of responses, SARS-CoV-2 infection primes the immune system with a broader range of antigens than vaccination. Anti-N seropositivity was strongly associated with protection against reported BIs in our cohort. This corroborates the conclusions of other studies, with previous estimates suggesting that it afforded participants with approximately 80% protection against SARS-CoV-2 re-infection for the subsequent 8-months 77. In those experiencing BIs, it is also associated with a shorter duration and reduced viral load 61. Evidence suggests that COVID-19 vaccination drives differential responses to subsequent SARS-CoV-2 infection, boosting those primed by the vaccine over new responses to the broader repertoire of proteins in the virus 61,78–80. Seroconversion to N upon SARS-CoV-2 infection was observed at a higher rate in placebo vaccinated participants (93%) compared to mRNA-1273 recipients (40%) 78. Additionally, SARS-CoV-2 BIs elicit anti-N IgG at significantly lower rates in infection-naïve individuals compared to those with a history of pre-vaccination COVID-19 61,79,80. This suggests that the order in which hybrid immunity is achieved may be important. SARS-CoV-2 LFT testing of a wider sample of ALSPAC participants highlighted an age-related difference in anti-N positivity rates. By spring 2022, all UK adults had been offered 3 vaccine doses, and this was reflected in comparable anti-S positivity rates of 93.0% and 96.0% in the G0/G1 cohorts respectively. However, only 50.3% of the older generation had a positive anti-N response compared to 67.1% of the younger adults. These values may underestimate SARS-CoV-2 infection rates in the older generation due to the lower N-seroconversion rates in infections occurring post-vaccination. However, they do accurately reflect a lack of potentially protective anti-N antibodies in this population. Prioritising older adults for early vaccination, and encouraging shielding behaviours, may therefore have consequences for the breadth and durability of the immunity they obtain by reducing pre-vaccination infection rates, and overall N-seroconversion. Rapid vaccination of this age group proved essential for reducing the rates of severe disease and associated mortality, and on a population level these benefits are unlikely to be outweighed by the benefits of a broader infection-primed response. Instead, this highlights the importance of designing novel vaccines that prime responses to additional antigens to generate more robust and sustained protection. Our BI susceptibility analysis should be interpreted in the context of the limitations of our study design. Firstly, the sample size was limited by those with available post-dose 2 vaccination measures and may not be representative of the whole population. Secondly, with respect to the BI rates in the case and controls groups, differences in exposure risk and test-seeking behaviours which could not be measured or accounted for may have influenced outcomes. Key variables, such as the proportion who received a booster vaccine, were however comparable between groups. Thirdly, N-protein serology does not offer 100% sensitivity for detection of new SARS-CoV-2 cases and therefore undetected infections during the study period could have resulted in participants being mis-classified as controls for the BI analysis. Fourthly, samples were not collected at the point of BI and which therefore could not be confirmed virologically, nor the SARS-CoV-2 sequences/variant lineages determined. The BI infection monitoring period covered the boundary of Delta and Omicron being the predominant variants in circulation. For the purposes of this analysis, all SARS-CoV-2 infections have been analysed together, but it is possible that variant-specific thresholds for protection may differ. Fifthly, although we report correlations between immune markers and susceptibility to future infection, we cannot exclude the possibility that rather than being directly protective, some may act as proxies for undetermined variables that weren’t measured. Finally, sample size limitations prevented the stratification of the BI analysis by vaccine type. However, this has previously been shown not to directly influence the susceptibility to BIs. Instead, higher BI rates in ChAdOx1 recipients can be attributed to the lower immunogenicity of ChAdOx1 resulting in participant IgG levels waning below protective thresholds faster than in BNT162b2 recipients 13. In summary, the data generated here add detail to the evidence of broader immune responses to infection and vaccination beyond the comparatively well-characterised anti-S IgG and neutralising antibody levels as markers of protection against SARS-CoV-2 BIs. Additionally, our findings support the notion that it is not only the magnitude of T-cell responses generated by hybrid immunity that improves protection against BI, but also their breadth and quality. This reinforces a need for next generation vaccines that elicit hybrid-mimicking immune responses for more robust and sustained protection. Better understanding the immune markers correlated with protection will also be important for the rapid validation of these new vaccines at a point in the pandemic when placebo-controlled vaccine trials are no longer practical. ## LAY SUMMARY FOR ALSPAC In this study we looked at immune responses to COVID-19 vaccination to try and understand how this may help predict whether someone is likely to have a SARS-CoV-2 infection in the future. All participants were part of the Children of the 90’s study and were aged from 27-70. Participants provided blood and saliva samples at three clinics (in December 2020, March 2021, and June 2021) which allowed us to monitor how their immune responses changed over time. We measured two different aspects of immunity - antibodies and T cells - that are both known to be important in protecting people from disease. Participants also completed questionnaires throughout the pandemic period to help us understand their SARS-CoV-2 infection history and provide details of their COVID-19 vaccinations. This study adds to, and supports, previous research showing that the type of immune response you generate from a combination of having a SARS-CoV-2 infection and being vaccinated against COVID-19, is different to your immune response if you just receive a vaccine. These results are important for understanding how scientists might design better vaccines against COVID-19 in future by mimicking the immunity you gain from an infection, without the associated negative consequences. ## Supporting information Supplementary Information [[supplements/308948_file02.pdf]](pending:yes) ## Data Availability All data produced in the present study are available upon reasonable request to the authors. ## FUNDING This work was funded by UK Research and Innovation (UKRI) and the National Institute of Health Research (NIHR) through the UK COVID-19 Immunology Consortium (UK CIC). This work was supported by the Elizabeth Blackwell Institute, University of Bristol, with funding from the Wellcome Trust ISSF3 grant 204813/Z/16/Z, and the University’s Alumni and Friends (LW, LR, AH, AD, AF and OF). NJT is the PI of the Avon Longitudinal Study of Parents and Children (MRC & WT 217065/Z/19/Z), is supported by the University of Bristol NIHR Biomedical Research Centre (BRC-1215-2001), the MRC Integrative Epidemiology Unit (MC_UU_00011/1) and works within the CRUK Integrative Cancer Epidemiology Programme (C18281/A29019). MK is supported by the Medical Research Council (MR/W021315/1). MS is supported by the Academy of Medical Sciences (Springboard Award SB007\100173). The UK Medical Research Council and the Wellcome Trust (Grant ref: 217065/Z/19/Z) and the University of Bristol provide core support for ALSPAC. This publication is the work of the authors; LR, AH, LW, HEB and MS will serve as guarantors for the contents of this paper. A comprehensive list of grant funding is available on the ALSPAC website ([http://www.bristol.ac.uk/alspac/external/documents/grant-acknowledgements.pdf](http://www.bristol.ac.uk/alspac/external/documents/grant-acknowledgements.pdf)). This research was specifically funded by Wellcome Trust and MRC grant 102215/2/13/2. The funders had no role in the study design, data collection, data analysis nor preparation of the manuscript or decision to publish. ## CONFLICTS OF INTEREST AF was a lead investigator on trials of COVID-19 vaccines funded by Oxford/Astra-Zeneca, Valneva and Sanofi and the UK government. He also leads a University of Bristol sponsored epidemiological study of adult respiratory disease funded by Pfizer which has evaluated COVID-19 vaccine effectiveness. During the pandemic he was a member of the Joint Committee on Vaccination and Immunisation which advised the UK government on COVID-19 vaccine policy and of the WHO Specialist Advisory Group of Experts COVID-19 vaccine working group. No other authors declare competing interests. ## ACKNOWLEDGEMENTS We are extremely grateful to all the families who took part in this study, the midwives for their help in recruiting them, and the whole ALSPAC team, which includes interviewers, computer and laboratory technicians, clerical workers, research scientists, volunteers, managers, receptionists and nurses. We wish to acknowledge the assistance of Dr. Andrew Herman, Helen Rice, and the University of Bristol Faculty of Health and Life Sciences Flow Cytometry Facility. This work was undertaken with support of the Elizabeth Blackwell Institute (EBI) Mechanisms to Populations Research Strand that promotes interdisciplinary research between fundamental bioscience and population health. The support from the EBI and the University’s Alumni and Friends funded the purchase of the CTL ELISpot reader and ELISA microplate washer which were used in this study. Thank you to the Bristol UNCOVER group for helpful discussions on assay development and the interpretation of results. * Received June 14, 2024. * Revision received June 14, 2024. * Accepted June 15, 2024. * © 2024, 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.Fink, Gunther, Tediosi, F. & Felder, S. Burden of Covid-19 restrictions: National, regional and global estimates. (2022) doi:10.1016/j. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j&link_type=DOI) 2. 2.Thompson, E. J. et al. Long COVID burden and risk factors in 10 UK longitudinal studies and electronic health records. Nat Commun 13, (2022). 3. 3.Richards, F., et al. Economic Burden of COVID-19: A Systematic Review. ClinicoEconomics and Outcomes Research vol. 14 293–307 Preprint at doi:10.2147/CEOR.S338225 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2147/CEOR.S338225&link_type=DOI) 4. 4.Wang, H. et al. Modelling the economic burden of SARS-CoV-2 infection in health care workers in four countries. Nat Commun 14, (2023). 5. 5.Hu, F. H. et al. Clinical outcomes of the severe acute respiratory syndrome coronavirus 2 Omicron and Delta variant: systematic review and meta-analysis of 33 studies covering 6 037 144 coronavirus disease 2019–positive patients. Clinical Microbiology and Infection vol. 29 835– 844 Preprint at doi:10.1016/j.cmi.2023.03.017 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cmi.2023.03.017&link_type=DOI) 6. 6.Hu, B., Guo, H., Zhou, P. & Shi, Z. L. Characteristics of SARS-CoV-2 and COVID-19. Nature Reviews Microbiology vol. 19 141–154 Preprint at doi:10.1038/s41579-020-00459-7 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41579-020-00459-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33024307&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 7. 7.Rahman, Md. O., et al. Protection of the third-dose and fourth-dose mRNA vaccines against SARS-CoV-2 Omicron subvariant: a systematic review and meta-analysis. BMJ Open 13, e076892 (2023). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYm1qb3BlbiI7czo1OiJyZXNpZCI7czoxMzoiMTMvMTIvZTA3Njg5MiI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzA2LzE1LzIwMjQuMDYuMTQuMjQzMDg5NDguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 8. 8.Feikin, D. R. et al. Duration of effectiveness of vaccines against SARS-CoV-2 infection and COVID-19 disease: results of a systematic review and meta-regression. The Lancet 399, 924–944 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/s0140-6736(22)00152-0&link_type=DOI) 9. 9.Wu, N. et al. Long-term effectiveness of COVID-19 vaccines against infections, hospitalisations, and mortality in adults: findings from a rapid living systematic evidence synthesis and meta-analysis up to December, 2022. Lancet Respir Med 11, 439–452 (2023). 10. 10.Desantis, S. M. et al. Incidence and Predictors of Breakthrough and Severe Breakthrough Infections of SARS-CoV-2 after Primary Series Vaccination in Adults: A Population-Based Survey of 22 575 Participants. Journal of Infectious Diseases 227, 1164–1172 (2023). 11. 11.Piñana, J. L. et al. One-year breakthrough SARS-CoV-2 infection and correlates of protection in fully vaccinated hematological patients. Blood Cancer J 13, (2023). 12. 12.Bergwerk, M. et al. Covid-19 Breakthrough Infections in Vaccinated Health Care Workers. New England Journal of Medicine 385, 1474–1484 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2109072&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 13. 13.Aldridge, R. W. et al. SARS-CoV-2 antibodies and breakthrough infections in the Virus Watch cohort. Nat Commun 13, (2022). 14. 14.Moss, P. The T cell immune response against SARS-CoV-2. Nature Immunology vol. 23 186–193 Preprint at doi:10.1038/s41590-021-01122-w (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41590-021-01122-w&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=35105982&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 15. 15.Ma, H. et al. Serum IgA, IgM, and IgG responses in COVID-19. Cellular and Molecular Immunology vol. 17 773–775 Preprint at doi:10.1038/s41423-020-0474-z (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41423-020-0474-z&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32467617&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 16. 16.Chia, W. N. et al. Dynamics of SARS-CoV-2 neutralising antibody responses and duration of immunity: a longitudinal study. Lancet Microbe 2, e240–e249 (2021). 17. 17.Montague, B. T. et al. Elevated serum IgA following vaccination against SARS-CoV-2 in a cohort of high-risk first responders. Sci Rep 12, (2022). 18. 18.Yang, Z. R. et al. Efficacy of SARS-CoV-2 vaccines and the dose–response relationship with three major antibodies: a systematic review and meta-analysis of randomised controlled trials. Lancet Microbe 4, e236–e246 (2023). 19. 19.Maringer, Y., et al. Durable Spike-Specific T Cell Responses after Different COVID-19 Vaccination Regimens Are Not Further Enhanced by Booster Vaccination. [https://www.science.org](https://www.science.org) (2022). 20. 20.Earle, K. A. et al. Evidence for antibody as a protective correlate for COVID-19 vaccines. Vaccine 39, 4423–4428 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.vaccine.2021.05.063&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 21. 21.Khoury, D. S. et al. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med 27, 1205–1211 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-021-01377-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 22. 22.Sheikh-Mohamed, S. et al. Systemic and mucosal IgA responses are variably induced in response to SARS-CoV-2 mRNA vaccination and are associated with protection against subsequent infection. Mucosal Immunol 15, 799–808 (2022). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 23. 23.Goh, Y. S. et al. Variant-Specific IgA Protects Against Omicron Infection. J Infect Dis (2023) doi:10.1093/infdis/jiad525. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/infdis/jiad525&link_type=DOI) 24. 24.Havervall, S. et al. Anti-Spike Mucosal IgA Protection against SARS-CoV-2 Omicron Infection. New England Journal of Medicine 387, 1333–1336 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/nejmc2209651&link_type=DOI) 25. 25.Zuo, J. et al. Robust SARS-CoV-2-specific T cell immunity is maintained at 6 months following primary infection. Nat Immunol 22, 620–626 (2021). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 26. 26.Hoffmann, M. et al. The Omicron variant is highly resistant against antibody-mediated neutralization: Implications for control of the COVID-19 pandemic. Cell 185, 447–456.e11 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/J.CELL.2021.12.032&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 27. 27.Planas, D. et al. Reduced sensitivity of SARS-CoV-2 variant Delta to antibody neutralization. Nature 596, 276–280 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1101/2021.05.26.445838&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 28. 28.Loyal, L. et al. Cross-reactive CD4+ T cells enhance SARS-CoV-2 immune responses upon infection and vaccination. Science (1979) 374, (2021). 29. 29.Swadling, L. et al. Pre-existing polymerase-specific T cells expand in abortive seronegative SARS-CoV-2. Nature 601, 110–117 (2022). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 30. 30.Northstone, K. et al. The Avon Longitudinal Study of Parents and children ALSPAC G0 Partners: A cohort profile. Wellcome Open Res 8, 37 (2023). 31. 31.Fraser, A., et al. Cohort profile: The avon longitudinal study of parents and children: ALSPAC mothers cohort. Int J Epidemiol 42, 97–110 (2013). 32. 32.Boyd, A. et al. Cohort profile: The ‘Children of the 90s’-The index offspring of the avon longitudinal study of parents and children. Int J Epidemiol 42, 111–127 (2013). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/ije/dys064&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22507743&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000316699300012&link_type=ISI) 33. 33.Northstone, K., et al. The Avon Longitudinal Study of Parents and Children (ALSPAC): an update on the enrolled sample of index children in 2019. Wellcome Open Res 4, (2019). 34. 34.ALSPAC data dictionary. [http://www.bristol.ac.uk/alspac/researchers/our-data/](http://www.bristol.ac.uk/alspac/researchers/our-data/). 35. 35.Halliday, A. et al. Development and evaluation of low-volume tests to detect and characterize antibodies to SARS-CoV-2. Front Immunol 13, (2022). 36. 36.Thomas, A. C. et al. Evaluation and deployment of isotype-specific salivary antibody assays for detecting previous SARS-CoV-2 infection in children and adults. Communications Medicine 3, 37 (2023). 37. 37.Harris, P. A. et al. Research electronic data capture (REDCap)-A metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform 42, 377–381 (2009). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jbi.2008.08.010&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18929686&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000264958800018&link_type=ISI) 38. 38.Payne, R. P. et al. Immunogenicity of standard and extended dosing intervals of BNT162b2 mRNA vaccine. Cell 184, 5699–5714.e11 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2021.10.011&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34735795&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 39. 39.Shrotri, M. et al. Spike-antibody responses to COVID-19 vaccination by demographic and clinical factors in a prospective community cohort study. Nat Commun 13, (2022). 40. 40.Sano, K. et al. SARS-CoV-2 vaccination induces mucosal antibody responses in previously infected individuals. Nat Commun 13, (2022). 41. 41.Azzi, L. et al. Mucosal immune response in BNT162b2 COVID-19 vaccine recipients. (2022) doi:10.1016/j. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j&link_type=DOI) 42. 42.Alu, A., Chen, L., Yuquan, H. L., Tian, W. X. & Wei, X. Intranasal COVID-19 vaccines: From bench to bed-NC-ND license ([http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/)). EBioMedicine 76, 103841 (2022). 43. 43.Mudd, P. A. et al. SARS-CoV-2 mRNA vaccination elicits a robust and persistent T follicular helper cell response in humans. Cell 185, 603–613.e15 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2021.12.026&link_type=DOI) 44. 44.Swanson Ii, P. A., et al. AZD1222/ChAdOx1 NCoV-19 Vaccination Induces a Polyfunctional Spike Protein-Specific T H 1 Response with a Diverse TCR Repertoire. Sci. Transl. Med vol. 13 [https://www.science.org](https://www.science.org) (2021). 45. 45.Maringer, Y., et al. Durable Spike-Specific T Cell Responses after Different COVID-19 Vaccination Regimens Are Not Further Enhanced by Booster Vaccination. [https://www.science.org](https://www.science.org) (2022). 46. 46.Rivino, L. et al. Virus-Specific T Lymphocytes Home to the Skin during Natural Dengue Infection. [https://www.science.org](https://www.science.org). 47. 47.Kim, T. S. & Shin, E. C. The activation of bystander CD8+ T cells and their roles in viral infection. Experimental and Molecular Medicine vol. 51 Preprint at doi:10.1038/s12276-019-0316-1 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s12276-019-0316-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31827070&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 48. 48.Messina, N. L., Zimmermann, P. & Curtis, N. The impact of vaccines on heterologous adaptive immunity. Clinical Microbiology and Infection vol. 25 1484–1493 Preprint at doi:10.1016/j.cmi.2019.02.016 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cmi.2019.02.016&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 49. 49.Lee, H. G., Cho, M. Z. & Choi, J. M. Bystander CD4+ T cells: crossroads between innate and adaptive immunity. Experimental and Molecular Medicine vol. 52 1255–1263 Preprint at doi:10.1038/s12276-020-00486-7 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s12276-020-00486-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32859954&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 50. 50.Kim, T. S. & Shin, E. C. The activation of bystander CD8+ T cells and their roles in viral infection. Experimental and Molecular Medicine vol. 51 Preprint at doi:10.1038/s12276-019-0316-1 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s12276-019-0316-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31827070&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 51. 51.Di Genova, G., Savelyeva, N., Suchacki, A., Thirdborough, S. M. & Stevenson, F. K. Bystander stimulation of activated CD4+ T cells of unrelated specificity following a booster vaccination with tetanus toxoid. Eur J Immunol 40, 976–985 (2010). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/eji.200940017&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20104490&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 52. 52.Whiteside, S. K., Snook, J. P., Williams, M. A. & Weis, J. J. Bystander T Cells: A Balancing Act of Friends and Foes. Trends in Immunology vol. 39 1021–1035 Preprint at doi:10.1016/j.it.2018.10.003 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.it.2018.10.003&link_type=DOI) 53. 53.Hall, V. et al. Protection against SARS-CoV-2 after Covid-19 Vaccination and Previous Infection. New England Journal of Medicine 386, 1207–1220 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2118691&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 54. 54.Levin, E. G. et al. Waning Immune Humoral Response to BNT162b2 Covid-19 Vaccine over 6 Months. New England Journal of Medicine 385, e84 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMOA2114583&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 55. 55.Pérez-Alós, L. et al. Modeling of waning immunity after SARS-CoV-2 vaccination and influencing factors. Nat Commun 13, 1614 (2022). 56. 56.Carabelli, A. M., et al. SARS-CoV-2 variant biology: immune escape, transmission and fitness. Nature Reviews Microbiology vol. 21 162–177 Preprint at doi:10.1038/s41579-022-00841-7 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41579-022-00841-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=36653446&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 57. 57.Lustig, Y. et al. Superior immunogenicity and effectiveness of the third compared to the second BNT162b2 vaccine dose. Nat Immunol 23, 940–946 (2022). 58. 58.Feng, S. et al. Correlates of protection against symptomatic and asymptomatic SARS-CoV-2 infection. Nat Med 27, 2032–2040 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591–021–01540-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 59. 59.Gilbert, P. B. et al. Immune correlates analysis of the mRNA-1273 COVID-19 vaccine efficacy clinical trial. Science (1979) 375, 43–50 (2022). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 60. 60.Regev-Yochay, G. et al. Correlates of protection against COVID-19 infection and intensity of symptomatic disease in vaccinated individuals exposed to SARS-CoV-2 in households in Israel (ICoFS): a prospective cohort study. Lancet Microbe 4, e309–e318 (2023). 61. 61.Gromowski, G. D. et al. Humoral immune responses associated with control of SARS-CoV-2 breakthrough infections in a vaccinated US military population. EBioMedicine 94, (2023). 62. 62.Painter, M. M. et al. Prior vaccination promotes early activation of memory T cells and enhances immune responses during SARS-CoV-2 breakthrough infection. Nat Immunol 24, 1711–1724 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41590-023-01613-y&link_type=DOI) 63. 63.Neale, I. et al. CD4+ and CD8+ T cells and antibodies are associated with protection against Delta vaccine breakthrough infection: a nested case-control study within the PITCH study. mBio 14, (2023). 64. 64.Scurr, M. J. et al. Magnitude of venous or capillary blood-derived SARS-CoV-2-specific T cell response determines COVID-19 immunity. Nat Commun 13, 5422 (2022). 65. 65.Brasu, N. et al. Memory CD8+ T cell diversity and B cell responses correlate with protection against SARS-CoV-2 following mRNA vaccination. Nat Immunol 23, 1445–1456 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41590-022-01313-z&link_type=DOI) 66. 66.Almendro-Vázquez, P. et al. Cellular and humoral immune responses and breakthrough infections after three SARS-CoV-2 mRNA vaccine doses. Front Immunol 13, (2022). 67. 67.Goldberg, Y. et al. Protection and Waning of Natural and Hybrid Immunity to SARS-CoV-2. New England Journal of Medicine 386, 2201–2212 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2118946&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=35613036&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 68. 68.Abu-Raddad, L. J. et al. Association of Prior SARS-CoV-2 Infection With Risk of Breakthrough Infection Following mRNA Vaccination in Qatar. JAMA 326, 1930–1939 (2021). 69. 69.Bowman, K. A. et al. Hybrid Immunity Shifts the Fc-Effector Quality of SARS-CoV-2 mRNA Vaccine-Induced Immunity. mBio 13, (2022). 70. 70.Rodda, L. B. et al. Imprinted SARS-CoV-2-specific memory lymphocytes define hybrid immunity. Cell 185, 1588–1601.e14 (2022). 71. 71.Carazo, S. et al. Protection against omicron (B.1.1.529) BA.2 reinfection conferred by primary omicron BA.1 or pre-omicron SARS-CoV-2 infection among health-care workers with and without mRNA vaccination: a test-negative case-control study. Lancet Infect Dis 23, 45–55 (2023). 72. 72.Reynolds, C. J. et al. Heterologous Infection and Vaccination Shapes Immunity against SARS-CoV-2 Variants. [https://www.science.org](https://www.science.org). 73. 73.Walls, A. C. et al. SARS-CoV-2 breakthrough infections elicit potent, broad, and durable neutralizing antibody responses. Cell 185, 872–880.e3 (2022). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 74. 74.Bertoletti, A., Le Bert, N., Qui, M. & Tan, A. T. SARS-CoV-2-specific T cells in infection and vaccination. Cellular and Molecular Immunology vol. 18 2307–2312 Preprint at doi:10.1038/s41423-021-00743-3 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41423-021-00743-3&link_type=DOI) 75. 75.Nguyen, T. H. O. et al. T Cells Targeting SARS-CoV-2: By Infection, Vaccination, and Against Future Variants. Frontiers in Medicine vol. 8 Preprint at doi:10.3389/fmed.2021.793102 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fmed.2021.793102&link_type=DOI) 76. 76.Lo Tartaro, D., et al. Detailed characterization of SARS-CoV-2-specific T and B cells after infection or heterologous vaccination. Front Immunol 14, (2023). 77. 77.Kohler, P. et al. Impact of baseline SARS-CoV-2 antibody status on syndromic surveillance and the risk of subsequent COVID-19—a prospective multicenter cohort study. BMC Med 19, (2021). 78. 78.Follmann, D. et al. Antinucleocapsid Antibodies After SARS-CoV-2 Infection in the Blinded Phase of the Randomized, Placebo-Controlled mRNA-1273 COVID-19 Vaccine Efficacy Clinical Trial. Ann Intern Med 175, 1258–1265 (2022). 79. 79.Allen, N. et al. Serological markers of SARS-CoV-2 infection; anti-nucleocapsid antibody positivity may not be the ideal marker of natural infection in vaccinated individuals. Journal of Infection 83, e9–e10 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jinf.2021.08.012&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34384812&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F06%2F15%2F2024.06.14.24308948.atom) 80. 80.Williams, D. M., et al. Establishing SARS-CoV-2 membrane protein-specific antibodies as a valuable serological target via high-content microscopy. iScience 26, (2023).