Abstract
We included measurement of pre-existing immunity to human endemic coronaviruses (HCoV) in a Phase I/II study of SARS-CoV-2 spike protein vaccine candidates. A Binding Antibody Multiplex Assay measured HCoV-specific IgG to the receptor binding domain or full-length spike of human coronaviruses HKU1, 229E, NL63, and OC43. We found no evidence for the impact of HCoV antibodies on neutralizing and binding antibody responses to the candidate SARS-CoV-2 vaccines.
Similar content being viewed by others
Introduction
Soon after the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there were prevailing concerns that prior vaccination might trigger immunopathological responses to the infection. This led to recommendations for monitoring the human phases of the clinical development of candidate vaccines1. Severe clinical outcomes following respiratory syncytial virus (RSV) infection in infants who had been vaccinated with a formalin-inactivated, whole-virus RSV vaccine motivated these concerns2. Later, non-clinical studies were able to reproduce the clinical phenotype in macaques in pursuit of the elucidation of relevant mechanisms3. Additional non-clinical observations of antibody-dependent enhancement of infection following vaccination with candidate SARS-CoV-14,5,6,7 or Middle East Respiratory Syndrome coronavirus (MERS)8 vaccines provided reasons for vigilance. Much of this preceding research focused on characteristics of the antigen design or vaccine construct and composition, and less on host factors.
Subsequent experience demonstrated that human infection with SARS-CoV-2 is associated with a spectrum of clinical outcomes, ranging from asymptomatic to fatal disease. This heterogeneity of clinical outcome following infection with SARS-CoV-2 is not fully accounted for by baseline health status or comorbid conditions9. More recently, emerging viral variants have been notable for increased efficiency of transmission with apparently lesser clinical severity of clinical infection10. Pre-existing immune responses have also been identified as a factor for differences in clinical outcomes11. By contrast, human populations have regular seasonal exposure to ‘common cold’ human endemic coronaviruses (HCoVs), which are notable for a less severe course of clinical illness12.
Pre-existing immunity to HCoVs could impact the response to SARS-CoV-2 vaccination in several ways. For example, boosting cross-reactive responses could augment viral clearance. Alternatively, boosting of pre-existing responses could focus immune responses on previously generated epitopes rather than novel epitopes that differ between the viruses13. Furthermore, antibody function that facilitates uptake by macrophages is hypothesized as a mechanism whereby pre-existing, non-neutralizing responses could lead to more severe manifestations of clinical illness following infection14. It was also not known whether pre-existing immune responses to endemic coronaviruses could impact immune responses to SARS-CoV-2 candidate vaccines.
Given these considerations, we included measurement of pre-existing binding responses to four human endemic coronaviruses in the first-in-human clinical evaluation of a SARS-CoV-2 spike protein vaccine candidate that was subsequently licensed as a booster vaccine15,16,17. We have previously evaluated the cellular immune response to this vaccine candidate, where we found little evidence of baseline cellular reactivity to SARS-CoV-2 spike protein or spike peptides among participants in this trial18. Here, we evaluate the impact of baseline HCoV antibody response magnitude and breadth on neutralizing and binding antibody responses to the candidate SARS-CoV-2 vaccine.
Results
Among 436 VAT00001 trial participants (99.3% of the safety analysis population) with available Binding Antibody Multiplex Assay (BAMA data), reactivity to four endemic coronaviruses was detected in all participants and did not differ by age group or dosing regimen [Fig. 1A, Supplementary Tables 2 and 3]. The distribution of the HCoV log10-transformed binding antibody responses at baseline (D01) shows considerable overlap across age range and dosing regimen [Fig. 1A]. Samples were tested at a 1:100 dilution factor, and the log10-transformed response magnitude (MFI) ranged from 2.3 to 4.5 (Supplementary Table 2), with a minimal percentage of sample/analyte combinations exhibiting responses above the linear range of the assay (123/3488 sample/analyte combinations (3.5%). There were modest levels of correlation of the binding antibody response magnitude between endemic coronaviruses, being generally strongest between the RBD and spike antibody response magnitude for a given endemic coronavirus, e.g. highest between OC43 RBD and OC43 spike than other HCoVs (Supplementary Fig. 2). Principal components analysis (PCA) revealed that there was no clustering of antibody responses among the endemic human coronaviruses (Fig. 1C).
Distribution of A human endemic coronavirus log 10 transformed binding antibody multiplex assay (BAMA) response magnitudes in baseline samples (D01) among VAT00001 trial participants, B fold-rise in neutralization titer and SARS-CoV-2 binding antibody concentration at D36 from baseline, stratified by age stratum and dosing schedule. Treatment assignments are shown for both panels, and C principal components analysis (PCA) plot of human endemic coronavirus response magnitude and treatment assignment. Ab antibody, D01 day 1, D36 day 36, HCoV human coronavirus, MFI mean fluorescence intensity, PC principal component, RBD receptor binding domain.
In linear regression models, we evaluated whether treatment assignment and the response magnitude of binding antibody responses at D01 to four endemic coronavirus RBD and full-length spike proteins were predictive of neutralizing antibody response to SARS-CoV-2, as assessed in the microneutralization assay. Among participants 18–49 years of age allocated to the two-dose cohort, treatment assignments were significant predictors of neutralizing antibody response at D36, but none of the binding antibody responses at baseline to endemic coronaviruses were significant predictors of neutralizing response (Table 1). The lack of predictive findings of the endemic coronaviruses held true for the two-dose cohort in the 50+ years of age stratum (Supplementary Table 4) and single-dose or two-dose cohorts evaluated at D22 (Supplementary Tables 5 and 6). The regression analyses for the age-dosing schedule—D22/D36 permutations (not shown)—were consistent with these findings. Combining antibody response magnitude across all spike assays or all RBD assays into a single measure of CoV spike or RBD breadth was also not predictive of neutralizing responses when accounting for treatment assignment (18–49 years two-dose cohort, p = 0.11 and p = 0.93, respectively). Scatter plots of D36 neutralizing response (log10 transformed fold rise of neutralizing response at D36 compared to baseline) versus the D0 BAMA response magnitude [log10 transformed breadth scores or mean fluorescence intensity (MFI)], stratified by age stratum and dosing schedule, further support the lack of meaningful effect of the prior endemic human coronavirus exposure on the neutralizing antibody response to vaccination (Fig. 1B).
There were no meaningful differences in baseline endemic coronavirus response magnitude among participants who achieved a four-fold rise in neutralizing titers by D36 compared to baseline. However, a slightly higher response magnitude to the betacoronavirus OC43 spike was observed among participants displaying a four-fold rise (4.06 vs. 4.12 log10 units, p = 0.018).
We further evaluated whether treatment assignment and D01 endemic coronavirus response magnitude were predictive of binding antibody response to SARS-CoV-2 in a validated GCN-4 enzyme-linked immunosorbent assay (ELISA). As for neutralizing responses, among participants 18–49 years of age allocated to the two-dose cohort, treatment assignments were significant predictors of binding antibody response at D36, but none of the endemic coronaviruses were significant predictors of binding antibody response (Table 2). The lack of predictive findings held true for the two-dose cohort in the 50+ years of age stratum and single-dose or two-dose cohorts evaluated at D22 (data not presented). In keeping with the neutralizing antibody analyses, breadth scores for cumulative HCoV spike or RBD response at baseline were not significant predictors of binding antibody response to SARS-CoV-2 post-baseline.
Discussion
We found that prior exposure to endemic alpha and beta-coronaviruses was universally detected among healthy adult volunteers in a Phase I/II trial conducted in eight clinical trial sites in the United States. There was no meaningful predictive association between pre-existing binding antibody responses to four endemic coronaviruses and either neutralizing or binding responses to SARS-CoV-2 following vaccination with stabilized prefusion SARS-CoV-2 spike trimer recombinant protein vaccine candidates. By contrast, the dose and schedule of these candidate formulations were meaningful predictors of the SARS-CoV-2 immune response in the trial volunteers, who were not previously exposed to SARS-CoV-2 and who did not exhibit antibody reactivity to SARS-CoV-2 at the baseline visit.
Our findings are similar to those observed in an occupational longitudinal cohort of individuals who received mRNA vaccinations19. In that study, while baseline endemic coronavirus antibody levels were correlated with SARS-CoV-2 antibody levels following infection, the authors did not find an association between baseline human common cold virus immunity and SARS-CoV-2 antibody levels post-vaccination. Crowley et al. also found that antibody responses to endemic HCoV were not significantly associated with vaccine responses20. The consistency of these findings despite near-universal prior exposure to HCoVs indicates that these highly immunogenic vaccines are not impacted by HCoVs-related immune imprinting as observed with infection. Our findings suggest that recent HCoV exposure, which strongly impacts vaccine response in murine models, may not be relevant to human vaccine response. However, it is important to note that the timing of exposure in the current trial volunteers is not known. One limitation of the current study is that we did not assess the HCoV antibody response magnitude following vaccination.
The theoretical possibility for suboptimal immune responses to vaccination, which could be associated with increased severity of respiratory disease upon infection with circulating SARS-CoV-2, was a significant concern in the initial stages of coronavirus disease 2019 (COVID-19) vaccine development. As exposure to human endemic coronaviruses is extremely common, the theoretical concern for immune imprinting to adversely affect the development of responses to SARS-CoV-2 vaccines was raised as a potential mechanism that could lead to vaccine-associated enhanced respiratory disease (VAERD)1,21,22. VAERD has not been observed in clinical development of multiple COVID-19 vaccines that achieved regulatory authorization, including the CoV2 dTM preS-AS03 vaccine, nor following widespread deployment in public vaccination campaigns23,24,25. In the present study, we found no meaningful evidence for an impact of prior HCoV exposure on measures of early immune response to vaccination with prefusion-stabilized trimer recombinant protein vaccine candidates. The current study provides an immunological view that prior exposure to endemic coronaviruses does not substantially impact the vaccine-elicited antibody responses.
Methods
Clinical trial
VAT00001 was a Phase I/II randomized, modified double-blind, placebo-controlled, first-in-human, dose-escalation trial. The trial design and interim results have been described in the prior publication26. In brief, participants were healthy volunteers without prior exposure to SARS-CoV-2, stratified by age (18–49 years of age, 50 and above). The vaccine formulations evaluated included one or two dose levels (1.3 or 2.6 μg) of a recombinant protein vaccine comprising the stabilized prefusion spike of the ancestral (WA-1) SARS-CoV-2 strain, delivered with AS03 (GSK) or AF03 (Sanofi) adjuvant or without adjuvant. The dosing schedules evaluated included a single administration (D01) or two doses separated by a 21-day interval (D01, D22).
Assay methods
The microneutralization assay (Global Clinical Immunology, Sanofi Vaccines, Swiftwater, PA) and GCN-4 binding antibody enzyme-linked immunosorbent assay (ELISA; Nexelis, Laval, QC) methods have been described elsewhere26,27. Blood samples obtained at baseline and post-vaccination (D01, D22, D36) were assessed in the above assays.
A qualified Binding Antibody Multiplex Assay (BAMA) was used to detect endemic coronavirus-specific IgG in serum from participants at baseline (D01) in the VAT00001 trial. The assay detects antibodies to the receptor binding domain (RBD) and to full-length spike of endemic human coronaviruses HKU1, 229E, NL63, and OC43. Both forms of CoV antigens were chosen since RBD exhibits lower homology across CoVs, whereas the full-length spike contains regions that are more conserved across CoVs20,28,29. Antigens included in the assay are detailed in Supplementary Table 1. The method for detecting HCoV-specific antibodies, described previously9, includes sample incubation with antigen-coated beads for 30 min (MagPlex beads, Luminex Corporation). NeutrAvidin-coated beads are bound to a biotinylated rabbit anti-6xhis antibody, which then captures His-tagged HCov proteins in BAMA diluent (PBS, 1% milk blotto (w/v), 5% normal goat serum, 0.05% Tween). Samples were tested at a single dilution of 1:100 in assay diluent, then added to the bound beads and then incubated for 30 min on an orbital plate shaker. Human IgG HCoV-specific antibodies are then detected using an anti-human IgG phycoerythrin (PE) labeled detection antibody (Southern Biotech). Plates containing the bound microspheres/antigen/antibody complex are read on a BioPlex 200 instrument to measure fluorescence intensity (FI). Duplicate wells are averaged to provide a mean fluorescence intensity (MFI) value. Background binding (no sample and no antigen controls) is subtracted from each sample well (Net MFI), which is then used for statistical analysis. The dynamic range of the Bioplex instrument is 2.0–4.5 (log10 transformed MFI). Assay controls included a titrated HCoV seropositive plasma sample (HCoV-PC55 plasma, kindly provided by Dr. Paul Goepfert (University of Alabama) and HCoV spike (AB711725 mAb, kindly provided by Dr. Kevin Saunders, Duke Human Vaccine Institute) as outlined in Supplementary Table 1. Additionally, an irrelevant human monoclonal antibody (7B2) was used as a negative control. Additional details regarding the specificity of the assay for endemic HCoV and the 7B2 negative control mAb binding are shown in Supplementary Fig. 1 (A and B), including the non-reactivity of a SARS-CoV-2-specific monoclonal antibody (AB712384 mAb, kindly provided by Drs. Kevin Saunders and Barton Haynes, Duke Human Vaccine Institute) to the endemic HCoV antigens.
The study was done in compliance with the International Conference on Harmonization guidelines for Good Clinical Practice and the principles of the Declaration of Helsinki. The protocol and amendments were approved by the applicable Independent Ethics Committees and Institutional Review Boards and the regulatory agency as per local regulations. Written informed consent was obtained from the participants before any study procedures were done26.
Statistical methods
We evaluated the RBD or spike HCoV antibody magnitudes at D01 and vaccine formulation as predictors of the D22 or D36 post-vaccination neutralizing antibody titer or binding antibody concentration using linear regression stratified by age (18–49, 50+) and dosing schedule (single dose, two doses). Additionally, we examined RBD and spike breadth scores as predictors by calculating the mean magnitude of the four HCoV RBDs or four HCoV spike proteins, respectively. Scores were calculated by first averaging the response magnitudes of the four individual antigens within each panel for each sample (i.e., mean of [HKU1, 229E, NL63, and OC43 RBDs] and mean of [HKU1, 229E, NL63, and OC43 spike proteins]). The resultant mean was then log10 transformed for analysis and plotting.
Statistical analyses were conducted using R v4.3.0. Principal component analysis (PCA) was conducted using the R function ‘prcomp,’ and a plot of PC1 and PC2 was generated to visualize potential outliers or clusters and the relationship between those RBD or spike HCoV antibodies plotted using the R package ggfortify. Correlations were evaluated using Spearman’s rank correlation. An alpha level of 0.05 was used to denote statistical significance. P-values were not adjusted for multiple comparisons.
Data availability
Qualified researchers can request access to patient-level data and related study documents, including the clinical study report, study protocol with any amendments, blank case report forms, statistical analysis plan, and dataset specifications. Patient-level data will be anonymized, and study documents will be redacted to protect the privacy of trial participants. Further details on Sanofi’s data sharing criteria, eligible studies, and process for requesting access can be found at https://vivli.org/.
References
Lambert, P. H. et al. Consensus summary report for CEPI/BC March 12–13, 2020 meeting: assessment of risk of disease enhancement with COVID-19 vaccines. Vaccine 38, 4783–4791 (2020).
Kim, H. W. et al. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am. J. Epidemiol. 89, 422–434 (1969).
De Swart, R. L. et al. Immunization of macaques with formalin-inactivated respiratory syncytial virus (RSV) induces interleukin-13-associated hypersensitivity to subsequent RSV infection. J. Virol. 76, 11561–11569 (2002).
Bolles, M. et al. A double-inactivated severe acute respiratory syndrome coronavirus vaccine provides incomplete protection in mice and induces increased eosinophilic proinflammatory pulmonary response upon challenge. J. Virol. 85, 12201–12215 (2011).
Channappanavar, R. & Perlman, S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. Semin. Immunopathol. 39, 529–539 (2017).
Perlman, S. & Dandekar, A. A. Immunopathogenesis of coronavirus infections: implications for SARS. Nat. Rev. Immunol. 5, 917–927 (2005).
Tseng, C. T. et al. Immunization with SARS coronavirus vaccines leads to pulmonary immunopathology on challenge with the SARS virus. PLoS ONE 7, e35421 (2012).
Agrawal, A. S. et al. Immunization with inactivated Middle East Respiratory Syndrome coronavirus vaccine leads to lung immunopathology on challenge with live virus. Hum. Vaccin. Immunother. 12, 2351–2356 (2016).
Schuster, D. J. et al. Lower SARS-CoV-2-specific humoral immunity in people living with HIV-1 recovered from nonhospitalized COVID-19. JCI Insight 7, e158402 (2022).
Adjei, S. et al. Mortality risk among patients hospitalized primarily for COVID-19 during the Omicron and Delta Variant pandemic periods—United States, April 2020–June 2022. MMWR Morb. Mortal. Wkly Rep. 71, 1182–1189 (2022).
Murray, S. M. et al. The impact of pre-existing cross-reactive immunity on SARS-CoV-2 infection and vaccine responses. Nat. Rev. Immunol. 23, 304–316 (2023).
Nickbakhsh, S. et al. Epidemiology of seasonal coronaviruses: establishing the context for the emergence of coronavirus disease 2019. J. Infect. Dis. 222, 17–25 (2020).
Schiepers, A. et al. Molecular fate-mapping of serum antibody responses to repeat immunization. Nature 615, 482–489 (2023).
Smatti, M. K., Al Thani, A. A. & Yassine, H. M. Viral-induced enhanced disease illness. Front. Microbiol. 9, 2991 (2018).
European Medicines Agency. EMA Recommends Approval of VidPrevtyn Beta as a COVID 19 Booster Vaccine. https://www.ema.europa.eu/en/news/ema-recommends-approval-vidprevtyn-beta-covid-19-booster-vaccine (2022).
de Bruyn, G. et al. Safety and immunogenicity of a variant-adapted SARS-CoV-2 recombinant protein vaccine with AS03 adjuvant as a booster in adults primed with authorized vaccines: a phase 3, parallel-group study. EClinicalMedicine 62, 102109 (2023).
Kirsebom, F. C. M. et al. Effectiveness of the Sanofi/GSK (VidPrevtyn Beta) and Pfizer-BioNTech (Comirnaty Original/Omicron BA.4-5) bivalent vaccines against hospitalisation in England. EClinicalMedicine 71, 102587 (2024).
De Rosa, S. C. et al. Whole-blood cytokine secretion assay as a high-throughput alternative for assessing the cell-mediated immunity profile after two doses of an adjuvanted SARS-CoV-2 recombinant protein vaccine candidate. Clin. Transl. Immunol. 11, e1360 (2022).
Lin, C. Y. et al. Pre-existing humoral immunity to human common cold coronaviruses negatively impacts the protective SARS-CoV-2 antibody response. Cell Host Microbe 30, 83–96.e84 (2022).
Crowley, A. R. et al. Boosting of cross-reactive antibodies to endemic coronaviruses by SARS-CoV-2 infection but not vaccination with stabilized spike. Elife 11, 1–24 (2022).
Food and Drug Administration. Development and Licensure of Vaccines to Prevent COVID-19: Guidance for Industry—FDA. https://www.fda.gov/media/139638/download (2023).
Munoz, F. M. et al. Vaccine-associated enhanced disease: case definition and guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 39, 3053–3066 (2021).
Baden, L. R. et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N. Engl. J. Med. 384, 403–416 (2021).
Dillard, J. A. et al. Adjuvant-dependent impact of inactivated SARS-CoV-2 vaccines during heterologous infection by a SARS-related coronavirus. Nat. Commun. 15, 3738 (2024).
Zhang, T. et al. Th2 and Th17-associated immunopathology following SARS-CoV-2 breakthrough infection in spike-vaccinated ACE2-humanized mice. J. Med. Virol. 96, e29408 (2024).
Goepfert, P. A. et al. Safety and immunogenicity of SARS-CoV-2 recombinant protein vaccine formulations in healthy adults: interim results of a randomised, placebo-controlled, phase 1-2, dose-ranging study. Lancet Infect. Dis. 21, 1257–1270 (2021).
Kalnin, K. V. et al. Immunogenicity and efficacy of mRNA COVID-19 vaccine MRT5500 in preclinical animal models. NPJ Vaccines 6, 61 (2021).
Premkumar, L. et al. The receptor binding domain of the viral spike protein is an immunodominant and highly specific target of antibodies in SARS-CoV-2 patients. Sci. Immunol. 5, 1–8 (2020).
Segovia-Chumbez, B., Graham, S. D., Jadi, R., de Silva, A. M. & Premkumar, L. Production of the receptor-binding domain of the viral spike proteins from 2003 and 2019 SARS CoVs and the Four common human coronaviruses for serologic assays and inhibitor screening. Bio Protoc. 11, e4026 (2021).
Acknowledgements
This study was funded by Sanofi and by federal funds from the Biomedical Advanced Research and Development Authority (BARDA), part of the office of the Assistant Secretary for Preparedness and Response at the US Department of Health and Human Services in collaboration with the US Department of Defense Joint Program Executive Office for Chemical, Biological, Radiological, and Nuclear Defense under contract number HHSO100201600005I. This work was done in collaboration with GlaxoSmithKline, which provided access to and use of the AS03 adjuvant system. The authors thank all participants, investigators, and study site personnel who took part in the study. The authors wish to thank Lode Schuerman and Marguerite Koutsoukos of GlaxoSmithKline for their contribution to this study. The authors also thank Michael Archibald, Judith Lucas, Saman Baral, and Tara McNair for expert technical assistance, Angelina Sharak and Jaiden Dumas for data processing, Dr. Marcella Sarzotti-Kelsoe and the Duke Quality Assurance for Vaccine Immunogenicity Programs (QADVIP) team for quality assurance oversight, and Hanson Geevarghese from Sanofi for scientific writing assistance and providing editorial assistance as well as manuscript coordination. We thank Dr. Paul Goepfert (University of Alabama) for the provision of specimens for assay development, and Drs. Kevin Saunders and Barton Haynes of the Duke Human Vaccine Institute for the provision of mAb reagents for assay development. Development of the endemic HCoV assay was funded by the Bill and Melinda Gates Foundation (OPP1210938) and the National Institutes of Health, National Institute of Allergy and Infectious Diseases (NIAID) (HVTN Laboratory Center, FHCC; UM1 AI068619).
Author information
Authors and Affiliations
Contributions
G.d.B. contributed to the design, data acquisition, and data analysis or interpretation of data. H.A. contributed substantially to the data acquisition and data analysis, or interpretation of data. C.B. contributed to performing binding antibody assays, review/QC of data, and participated in data analysis. Y.J. contributed to data analysis or interpretation of data. L.P. contributed to the resources of the manuscript. S.V.M. participated in data interpretation and program management. S.S. contributed to data processing and binding antibody assay method qualification analysis. D.Z. contributed to data analysis or interpretation of data. R.M.C. contributed to the conception and study design. S.S. contributed to conception and design. G.D.T. contributed to study design and conceptualization, method development, supervision, and analysis. K.E.S. contributed to study design and conceptualization, methods development, supervision and analysis. All authors reviewed and approved the final manuscript.
Corresponding author
Ethics declarations
Competing interests
G.d.B. is an employee of Sanofi and received all support for this manuscript from Sanofi. He is also an inventor on a pending patent application filed by Sanofi and GSK for the development of the CoV-2 dTM vaccine. Additionally, he owns stock or stock options in Sanofi. Funding for this work was provided by the US Government through the Biomedical Advanced Research and Development Authority (BARDA) under contract HHSO100201600005I. H.A. received support for attending meetings and/or travel and holds stocks as an employee of Sanofi. L.P. received a US NIH grant to his institution under 4U54CA260543. C.B., S.S., S.V.M., G.T., and K.S. received funding and samples used for the analysis for this work at Duke University (PI Georgia Tomaras) from Sanofi. R.M.C. is an employee of Sanofi and holds patents and stocks of Sanofi. He also received a BARDA contract award from Sanofi. S.S. is an employee of Sanofi and holds stock and stock options in Sanofi. G.D.T. also notes NIH grants to her institution and funding from Regeneron and Janssen to her institution. Y.J. and D.Z. have no competing interests to declare.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
de Bruyn, G., Adhikarla, H., Brackett, C.K. et al. Prior human endemic coronavirus exposure does not affect humoral responses to SARS-CoV-2 protein vaccines. npj Vaccines 10, 153 (2025). https://doi.org/10.1038/s41541-025-01203-z
Received:
Accepted:
Published:
Version of record:
DOI: https://doi.org/10.1038/s41541-025-01203-z



