Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Correlates of protection against symptomatic SARS-CoV-2 in vaccinated children

Abstract

The paucity of information on longevity of vaccine-induced immune responses and uncertainty of the correlates of protection hinder the development of evidence-based COVID-19 vaccination policies for new birth cohorts. Here, to address these knowledge gaps, we conducted a cohort study of healthy 5–12-year-olds vaccinated with BNT162b2. We serially measured binding and neutralizing antibody titers (nAbs), spike-specific memory B cell (MBC) and spike-reactive T cell responses over 1 year. We found that children mounted antibody, MBC and T cell responses after two doses of BNT162b2, with higher antibody and T cell responses than adults 6 months after vaccination. A booster (third) dose only improved antibody titers without impacting MBC and T cell responses. Among children with hybrid immunity, nAbs and T cell responses were highest in those infected after two vaccine doses. Binding IgG titers, MBC and T cell responses were predictive, with T cells being the most important predictor of protection against symptomatic infection before hybrid immunity; nAbs only correlated with protection after hybrid immunity. The stable MBC and T cell responses over time suggest sustained protection against symptomatic SARS-CoV-2 infection, even when nAbs wane. Booster vaccinations do not confer additional immunological protection to healthy children.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Adaptive immune responses following BNT162b2 vaccination in children aged 5–12 years were comparable or superior to adults despite reduced vaccine dose.
Fig. 2: A booster dose increased antibody titers but had no effect on memory B cell and T cell responses for SARS-CoV-2-naive children.
Fig. 3: Superiority of the adaptive immune parameters in children who acquired hybrid immunity within 6 months post-vaccination compared to vaccination or infection alone.
Fig. 4: A booster dose had minimal effects on adaptive immune responses in children with hybrid immunity.
Fig. 5: Anti-S IgG S+ MBCs and T cell responses are correlates of protection against symptomatic SARS-CoV-2 infection before hybrid immunity.
Fig. 6: nAb titers correlate with protection against symptomatic SARS-CoV-2 infection when prevalence of hybrid immunity was high.

Similar content being viewed by others

Data availability

All aggregate data supporting the findings of this study are available within the paper and its supplementary materials. Individual-level participant data are not publicly available. Only the data of individuals who consented to further research can be accessible with the consent of the ethics committees from the requestor’s and corresponding authors’ institutions. A formal data transfer agreement between the institutions will be required upon ethics approval. The corresponding authors can be contacted for access to data and will respond within 1 month; data transfer can take place once the data transfer agreement is completed.

References

  1. World Health Organization. Interim statement on hybrid immunity and increasing population seroprevalence rates. https://www.who.int/news/item/01-06-2022-interim-statement-on-hybrid-immunity-and-increasing-population-seroprevalence-rates (2022).

  2. Goldblatt, D., Alter, G., Crotty, S. & Plotkin, S. A. Correlates of protection against SARS-CoV-2 infection and COVID-19 disease. Immunol. Rev. 310, 6–26 (2022).

    Article  CAS  PubMed  Google Scholar 

  3. 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).

    Article  CAS  PubMed  Google Scholar 

  4. Wherry, E. J. & Barouch, D. H. T cell immunity to COVID-19 vaccines. Science 377, 821–822 (2022).

    Article  CAS  PubMed  Google Scholar 

  5. Bertoletti, A., Le Bert, N. & Tan, A. T. SARS-CoV-2-specific T cells in the changing landscape of the COVID-19 pandemic. Immunity 55, 1764–1778 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kent, S. J. et al. Disentangling the relative importance of T cell responses in COVID-19: leading actors or supporting cast? Nat. Rev. Immunol. 22, 387–397 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kalimuddin, S. et al. Early T cell and binding antibody responses are associated with COVID-19 RNA vaccine efficacy onset. Medicine 2, 682–688 (2021).

    Article  CAS  Google Scholar 

  8. Tan, A. T. et al. Rapid measurement of SARS-CoV-2 spike T cells in whole blood from vaccinated and naturally infected individuals. J. Clin. Investig. 131, e152379 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Samandari, T. et al. Prevalence and functional profile of SARS-CoV-2 T cells in asymptomatic Kenyan adults. J. Clin. Investig. 133, e170011 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Le Bert, N. et al. Highly functional virus-specific cellular immune response in asymptomatic SARS-CoV-2 infection. J. Exp. Med. 218, e20202617 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Ogbe, A. et al. T cell assays differentiate clinical and subclinical SARS-CoV-2 infections from cross-reactive antiviral responses. Nat. Commun. 12, 2055 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sette, A. & Crotty, S. Immunological memory to SARS-CoV-2 infection and COVID-19 vaccines. Immunol. Rev. 310, 27–46 (2022).

    Article  CAS  PubMed  Google Scholar 

  13. Andreano, E. et al. Hybrid immunity improves B cells and antibodies against SARS-CoV-2 variants. Nature 600, 530–535 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Suryawanshi, R. & Ott, M. SARS-CoV-2 hybrid immunity: silver bullet or silver lining? Nat. Rev. Immunol. 22, 591–592 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Centers for Disease Control and Prevention. COVID-19 vaccine: interim COVID-19 immunization schedule for persons 6 months of age and older. https://broadwaymedicalclinic.com/wp-content/uploads/2020/04/covid-19-immunization-schedule-ages-6months-older.pdf (2023).

  16. Royal College of Paediatrics and Child Health. RCPCH statement in response to JCVI advice on offering COVID-19 vaccination to all 5–11-year-olds. https://www.rcpch.ac.uk/news-events/news/rcpch-statement-response-jcvi-advice-offering-covid-19-vaccination-all-5-11-year (2022).

  17. European Medicines Agency. Comirnaty. https://www.ema.europa.eu/en/medicines/human/EPAR/comirnaty (2021).

  18. Ministry of Health Singapore. Child vaccination. https://www.moh.gov.sg/covid-19/vaccination/child (2021).

  19. Feng, S. et al. Correlates of protection against symptomatic and asymptomatic SARS-CoV-2 infection. Nat. Med. 27, 2032–2040 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Corbett, K. S. et al. Immune correlates of protection by mRNA-1273 vaccine against SARS-CoV-2 in nonhuman primates. Science 373, eabj0299 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Earle, K. A. et al. Evidence for antibody as a protective correlate for COVID-19 vaccines. Vaccine 39, 4423–4428 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Khoury, D. S. et al. Predicting the efficacy of variant-modified COVID-19 vaccine boosters. Nat. Med. 29, 574–578 (2023).

    Article  CAS  PubMed  Google Scholar 

  23. Cromer, D. et al. Neutralising antibody titres as predictors of protection against SARS-COV-2 variants and the impact of boosting: a meta-analysis. Lancet Microbe https://doi.org/10.1016/s2666-5247(21)00267-6 (2022).

  24. Gilbert, P. B. et al. Immune correlates analysis of the mRNA-1273 COVID-19 vaccine efficacy clinical trial. Science 375, 43–50 (2022).

    Article  CAS  PubMed  Google Scholar 

  25. Dowell, A. C. et al. Association of spike-specific T cells with relative protection from subsequent SARS-CoV-2 Omicron infection in young children. JAMA Pediatr. 177, 96–97 (2023).

    Article  PubMed  Google Scholar 

  26. Tan, A. T. et al. Early induction of functional SARS-CoV-2-specific T cells associates with rapid viral clearance and mild disease in COVID-19 patients. Cell Rep. 34, 108728 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Rydyznski Moderbacher, C. et al. Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity. Cell 183, 996–1012 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bertoletti, A. et al. SARS-CoV-2-specific T cells in infection and vaccination. Cell Mol. Immunol. 18, 2307–2312 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Carabelli, A. M. et al. SARS-CoV-2 variant biology: immune escape, transmission and fitness. Nat. Rev. Microbiol. 21, 162–177 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. 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).

    Article  CAS  PubMed  Google Scholar 

  31. Crotty, S. Hybrid immunity. Science 372, 1392–1393 (2021).

    Article  CAS  Google Scholar 

  32. Goel, R. R. et al. Distinct antibody and memory B cell responses in SARS-CoV-2 naïve and recovered individuals following mRNA vaccination. Sci. Immunol. 6, eabi6950 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Goel, R. R. et al. mRNA vaccines induce durable immune memory to SARS-CoV-2 and variants of concern. Science 374, abm0829 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Yung, C. F. et al. BNT162b2 vaccine protection against omicron and effect of previous infection variant and vaccination sequence among children and adolescents in Singapore: a population-based cohort study. Lancet Child Adolesc. Health 7, 463–470 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Plotkin, S. A. Correlates of protection induced by vaccination. Clin. Vaccin. Immunol. 17, 1055–1065 (2010).

    Article  CAS  Google Scholar 

  36. Moss, P. The T cell immune response against SARS-CoV-2. Nat. Immunol. 23, 186–193 (2022).

    Article  CAS  PubMed  Google Scholar 

  37. McMahan, K. et al. Correlates of protection against SARS-CoV-2 in rhesus macaques. Nature 590, 630–634 (2021).

    Article  CAS  PubMed  Google Scholar 

  38. Madelon, N. et al. Robust T-cell responses in anti-CD20-treated patients following COVID-19 vaccination: a prospective cohort study. Clin. Infect. Dis. 75, e1037–e1045 (2022).

    Article  PubMed  Google Scholar 

  39. Md Yusof, M. Y. et al. Breakthrough SARS-CoV-2 infections and prediction of moderate-to-severe outcomes during rituximab therapy in patients with rheumatic and musculoskeletal diseases in the UK: a single-centre cohort study. Lancet Rheumatol. 5, e88–e98 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Cho, A. et al. Anti-SARS-CoV-2 receptor-binding domain antibody evolution after mRNA vaccination. Nature 600, 517–522 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Mateus, J. et al. Low-dose mRNA-1273 COVID-19 vaccine generates durable memory enhanced by cross-reactive T cells. Science 374, eabj9853 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tulsian, N. K. et al. Defining neutralization and allostery by antibodies against COVID-19 variants. Nat. Commun. https://doi.org/10.1038/s41467-023-42408-x (2023).

  44. Tan, C. W. et al. A SARS-CoV-2 surrogate virus neutralization test based on antibody-mediated blockage of ACE2-spike protein-protein interaction. Nat. Biotechnol. 38, 1073–1078 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Tan, C. W. et al. Pan-sarbecovirus neutralizing antibodies in BNT162b2-immunized SARS-CoV-1 survivors. N. Engl. J. Med. 385, 1401–1406 (2021).

    Article  CAS  PubMed  Google Scholar 

  46. Tan, C. W. et al. Comparative neutralisation profile of SARS-CoV-2 Omicron subvariants BA.2.75 and BA.5. Lancet Microbe 3, e898 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Gaebler, C. et al. Evolution of antibody immunity to SARS-CoV-2. Nature 591, 639–644 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sokal, A. et al. Maturation and persistence of the anti-SARS-CoV-2 memory B cell response. Cell 184, 1201–1213 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wang, Z. et al. mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants. Nature 592, 616–622 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We gratefully acknowledge the support from A. Bertoletti and A. Tanoto Tan for suggestions on data interpretation; E. Shuyi Gan, T. Siriphanitchakorn, Y. S. Leong, K. W. Teng, C. Qui and M. Qui from Duke-NUS and C.-H. Huang, G. C. Yap, H. Wen, B. Shunmuganathan and R. Gupta from NUS for technical assistance; clinical research coordinators S. Wong, J. Lim, J. Yap, R. Chua and N. Siti Binte Roslan from NUH for assistance with participant recruitment and follow-up; and S. Nishanti Ramasamy for assistance with manuscript editing, formatting and submission. E.H.T. is supported by the National Medical Research Council (NMRC) Transition Award (MOH-000269), C.W.T. is supported by the NMRC Open Fund – Large Collaborative Grant (OFLCG19May-0034) and the National University of Singapore Startup grant (NUHSRO/2023/018/Startup/10) and E.E.O. is supported by the NMRC Singapore Translational Research (STaR) Award (MOH-001271-00). This study was supported by the NUS award of E.H.T. (NUHSRO/2021/081/NUS Med/07/MARVELS) from the Yong Loo Lin School of Medicine. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. Most of all, we gratefully acknowledge the heroic efforts of all the children and families who gave their precious time and biosamples to the MARVELS study.

Author information

Authors and Affiliations

Authors

Contributions

Y.Z., L.P.S., E.H.T. and E.E.O. designed and conceptualized the pediatric MARVELS study. J.G.L. designed and conceptualized the adult healthcare worker study used for comparison. L.P.S. and E.H.T. acquired funding and supervised the project. A.Y.H.K., C.J.X.T., H.E.L., N.E., W.C.Y., C.W.T., J.M.E.L. and N.L.B. provided administrative, technical and material support. Y.Z. and C.J.X.T. performed the statistical analyses. Y.Z., K.R.C., E.Z.O. and E.E.O. interpreted the data. Y.Z. and E.E.O. wrote the paper. All authors contributed to the revision of the paper and approved the final version for publication.

Corresponding authors

Correspondence to Youjia Zhong or Eng Eong Ooi.

Ethics declarations

Competing interests

N.L.B. reports a patent for a method to monitor SARS-CoV-2-specific T cells in biological samples, which is pending. The other authors declare no competing interests.

Peer review

Peer review information

Nature Medicine thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Alison Farrell, in collaboration with the Nature Medicine team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Isotypes and binding activity of Spike-specific memory B cells (S + MBC) in children with immunity against SARS-CoV-2.

A. Representative flow cytometric staining plots for vaccine-only (VV), hybrid (VVI), and infection-only immunity (n = 1 each). B. S + MBC isotypes in children with vaccine-only immunity (n = 34), at pre-vaccination baseline, and 3- and 6 months post vaccination. C. Binding activity against Spike proteins from three SARS-CoV-2 variants in B cell ELISPOT. Spike-specific memory B cells (S+ MBCs) were extracted from a pediatric subject 3 months after 2 doses of mRNA BNT162b2, and then cultured and differentiated ex vivo into antibody secreting cells.

Extended Data Fig. 2 Children vaccinated with 2 doses of mRNA SARS-CoV-2 vaccine have low levels of Th2 cytokines (n = 34).

A, B, C. Spike-reactive Th2 responses, measured by interleukin-4, interleukin-5 and interleukin-13 for children with vaccine-only immunity, and adults. For box-whisker graphs, upper and lower boundaries of boxes indicate upper and lower quartile respectively, line indicates median, and whiskers represent the range.

Extended Data Fig. 3 Vaccine-associated local and systemic adverse events and symptoms of SARS-CoV-2 infection reported by MARVELS children.

A. Percentages and severity of parent and / or subject – reported local and systemic symptoms for 10 days after vaccination. Blue = adverse events after doses 1 and 2, with color gradient representing spectrum of severity; Red = adverse events after booster (dose 3), with color gradient representing spectrum of severity. B. Clinical features of symptomatic SARS-CoV-2 infections for all episodes of symptomatic SARS-CoV-2 infection among vaccinated children. Two-tailed Fisher’s exact test was used for comparison between groups; ns: not significant, *: p ≤ 0.05, **: p ≤ 0.01, ***: p ≤ 0.001, ****: p ≤ 0.0001. For pain at injection site, p-value = 0.0189. For fever, p-value = 0.0091.

Extended Data Fig. 4 Nucleocapsid (N)-specific antibodies and T cells in children with asymptomatic SARS-CoV-2 infections.

A. Venn diagram showing all episodes of asymptomatic COVID-19 identified in this study (n = 30), separated into whether they were identified via seropositivity, T cell reactivity, or both against SARS-CoV-2 N protein. B, C. N-reactive T cell responses measured by post-stimulation interferon-γ and interleukin-2 levels, at pre-vaccination baseline, 3- and 6 months post vaccination, from vaccinated (two doses) children who remained uninfected (VV, n = 34), with symptomatic infection (VVI(S), n = 49) and asymptomatic infection (VVI(A), n = 11). D. N-reactive T cell responses measured by post-stimulation interferon-γ and E. interleukin-2 levels, at 6- and 12 months post vaccination, for VV children at 6 months. F. N-reactive T cell responses measured by post-stimulation interferon-γ and G. interleukin-2 levels, at 6- and 12 months post vaccination, for children with hybrid immunity at 6 months. In D-G, children were grouped into the following categories: 1) Those who did not develop SARS-CoV-2 infection between 6 and 12 months, 2) those who developed symptomatic SARS-CoV-2 re-infection (S), or 3) those who had asymptomatic re-infection. In all graphs, subjects with serological evidence of asymptomatic SARS-CoV-2 are colored in blue.

Extended Data Fig. 5 The proximity of last immunity-boosting event is similar for children who had symptomatic SARS-CoV-2 infection between doses 1 and 2 (VIV), and children who had symptomatic SARS-CoV-2 infection after 2 doses (VVI(S)).

A. Schematic representation of the different last immunity-boosting event, for VIV and VVI(S) children. Created with BioRender.com. B. Comparison of proximity of immunity-boosting event for VIV and VVI(S) children. For box-whisker graphs, upper and lower boundaries of boxes indicate upper and lower quartile respectively, line indicates median, and whiskers represent the range. Two-tailed Mann–Whitney U test was used for comparison between groups; ns: not significant, *: p ≤ 0.05, **: p ≤ 0.01, ***: p ≤ 0.001, ****: p ≤ 0.0001.

Extended Data Fig. 6 Eight-cytokines measured in the cytokine release assay for Spike-reactive T cell responses among children with hybrid immunity.

A. Schematic of cytokine release assay and analytical methods (after DMSO control subtraction) using unsupervised clustering algorithm (UMAP). The cytokines quantified were interferon-γ (IFN-γ), interleukin-2 (IL-2), tumor necrosis factor-α (TNF-α), Granzyme-B, interleukin-10 (IL-10), interleukin-4 (IL-4), interleukin-5 (IL-5), and interleukin-13 (IL-13). B. UMAP plots generated with all analyzed samples (n = 391, consisting of adults, infected children and vaccinated children at all time points) with levels of secreted cytokines shown in heatmaps. C. Concatenated cytokine secretion profiles of S peptide pool-stimulated whole blood from children with VIV, VVI(S) and VVI(A). Cytokine secretion profiles were overlaid on the global UMAP plot of all analyzed samples (black dots; each dot corresponds to one culture supernatant).

Extended Data Fig. 7 Immune correlates of protection against symptomatic SARS-CoV-2 infection between 3- and 6 months from start of vaccination.

N = 23 children had symptomatic SARS-CoV-2 infection between months 3 and 6, while n = 66 did not; of these, n = 3 had asymptomatic SARS-CoV-2 infection. A. 3-month Spike (S)-reactive T cell responses, quantified using interferon- γ (IFN-γ) and interleukin-2 (IL-2), for children with no infection, symptomatic and asymptomatic SARS-CoV-2 infection between 3 and 6 months. For comparison between no infection and symptomatic infection, p-value for IFN-γ = 0.000688 and for IL-2 = 0.025786. For comparison between symptomatic infection and asymptomatic infection, p-value for IFN-γ = 0.015810 and for IL-2 = 0.031621. B. 3-month Omicron BA.2 pVNT50 titers in children who did and did not develop symptomatic SARS-CoV-2 infection between 3 and 6 months. Color of dots indicate the type of pre-existing immunity they had at month 3 (p-value = 0.000589). C. Receiver operator characteristics (ROC) curve for BA.2 measured at 3 months from start of vaccination. D. pVNT50 titers Anti-S IgG titers, E. Percentage of S+ MBCs out of total B cells (p-value 0.004599), and F. S-reactive T cell responses measured by post-stimulation interferon-γ levels at month 3 post-vaccination (p-value 0.047200), for VVI(S) and non-VVI(S) children with and without symptomatic SARS-CoV-2 infection. For box-whisker graphs, upper and lower boundaries of boxes indicate upper and lower quartile respectively, line indicates median, and whiskers represent the range. Two-tailed Mann–Whitney U test was used to compare between groups. The ROC curve analysis was performed using the Wilson/Brown test. ns: not significant, *: p ≤ 0.05, **: p ≤ 0.01, ***: p ≤ 0.001, ****: p ≤ 0.0001.

Extended Data Fig. 8 Protection from symptomatic SARS-CoV-2 infection was associated with hybrid immunity but not age.

A. Superior protective capacity of hybrid immunity, demonstrated by the different in percentages of children who developed symptomatic SARS-CoV-2 infection between 6 and 12 months, among children with vaccine-only (VV) immunity at 6 months, compared to children with hybrid immunity at 6 months. Two-tailed Fisher’s exact test was used for comparison between groups. P-value = 0.0018. B, C. Symptomatic SARS-CoV-2 infection was not associated with age, as demonstrated by B. age distribution of all children getting their first infection after 2 doses of mRNA vaccination (n = 57 symptomatic, n = 16 asymptomatic), and C. age distribution of all children with hybrid immunity getting a re-infection (n = 3 symptomatic and n = 11 asymptomatic). For box-whisker graphs, upper and lower boundaries of boxes indicate upper and lower quartile respectively, line indicates median, and whiskers represent the range. Two-tailed Mann–Whitney U test was used to compare between different groups. ns: not significant, *: p ≤ 0.05, **: p ≤ 0.01, ***: p ≤ 0.001, ****: p ≤ 0.0001.

Extended Data Table 1 Baseline demographic and clinical features of all pediatric subjects
Extended Data Table 2 Multivariate regression model of the humoral and cellular immune parameters and their correlation with symptomatic SARS-CoV-2 infection between 3 and 6 months post-vaccination (dependent variable)

Supplementary information

Supplementary Information

Supplementary Fig. 1 and Table 1.

Reporting Summary

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhong, Y., Kang, A.Y.H., Tay, C.J.X. et al. Correlates of protection against symptomatic SARS-CoV-2 in vaccinated children. Nat Med (2024). https://doi.org/10.1038/s41591-024-02962-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41591-024-02962-3

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing