51
|
Izac JR, Kwee EJ, Gaigalas A, Wang L. Quantitative and Standardized Pseudovirus Neutralization Assay for COVID-19. Methods Mol Biol 2024; 2779:259-271. [PMID: 38526789 DOI: 10.1007/978-1-0716-3738-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
COVID-19 is a global pandemic caused by the highly infectious SARS-CoV-2 virus. Efforts to combat SARS-CoV-2 infection include mass vaccination and development of monoclonal and convalescent plasma therapeutics that require precise measurements of correlative, functional neutralizing antibodies that prevent virus infection. Developing rapid, safe, easy-to-use, and high-quality neutralization assays are essential for the success of the massive effort. Here, we developed a vesicular stomatitis virus-based neutralization assay that was capable of quantifying varying degrees of neutralization in patient serum samples. This assay has two detection readouts, flow cytometry and live cell imaging. The two readout methods produced consistent values of all 50% neutralization titers, further enhancing measurement confidence on the assay. Moreover, the use of available reference standards such as the World Health Organization International Standard (NIBSC code 20/136) enables quantification and standardization of the pseudovirus neutralization assay with neutralizing antibody titers measured in International Units/mL. Quantitative and standardized neutralization assays are critical for reliable efficacy evaluation and comparison of numerous vaccines and therapeutics.
Collapse
Affiliation(s)
- Jerilyn R Izac
- Biosystem and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA
| | - Edward J Kwee
- Biosystem and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA
| | - Adolfas Gaigalas
- Biosystem and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA
| | - Lili Wang
- Biosystem and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA.
| |
Collapse
|
52
|
Lovell JF, Miura K, Baik YO, Lee C, Lee JY, Park YS, Hong I, Lee JH, Kim T, Seo SH, Kim JO, Song M, Kim CJ, Choi JK, Kim J, Choo EJ, Choi JH. One-year antibody durability induced by EuCorVac-19, a liposome-displayed COVID-19 receptor binding domain subunit vaccine, in healthy Korean subjects. Int J Infect Dis 2024; 138:73-80. [PMID: 37944586 DOI: 10.1016/j.ijid.2023.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
OBJECTIVE EuCorVac-19 (ECV-19), an adjuvanted liposome-displayed receptor binding domain (RBD) COVID-19 vaccine, previously reported interim Phase 2 trial results showing induction of neutralizing antibodies 3 weeks after prime-boost immunization. The objective of this study was to determine the longer-term antibody response of the vaccine. METHODS To assess immunogenicity 6 and 12 months after vaccination, participants in the Phase 2 trial (NCT04783311) were excluded if they: 1) withdrew, 2) reported COVID-19 infection or additional vaccination, or 3) exhibited increasing Spike (S) antibodies (representing possible non-reported infection). Following exclusions, of the 197 initial subjects, anti-S IgG antibodies and neutralizing antibodies were further assessed in 124 subjects at the 6-month timepoint, and 36 subjects at the 12-month timepoint. RESULTS Median anti-S antibody half-life was 52 days (interquartile range [IQR]:42-70), in the "early" period from 3 weeks to 6 months, and 130 days (IQR:97-169) in the "late" period from 6 to 12 months. There was a negative correlation between initial antibody titer and half-life. Anti-S and neutralizing antibody responses were correlated. Neutralizing antibody responses showed longer half-lives; the early period had a median half-life of 120 days (IQR:81-207), and the late period had a median half-life of 214 days (IQR:140-550). CONCLUSION These data establish antibody durability of ECV-19, using a framework to analyze COVID-19 vaccine-induced antibodies during periods of high infection.
Collapse
Affiliation(s)
- Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, Buffalo, New York, USA.
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Yeong Ok Baik
- Eubiologics, R&D Center, EuBiologics Co., Ltd., Chuncheon, Korea
| | - Chankyu Lee
- Eubiologics, R&D Center, EuBiologics Co., Ltd., Chuncheon, Korea
| | - Jeong-Yoon Lee
- Eubiologics, R&D Center, EuBiologics Co., Ltd., Chuncheon, Korea
| | | | - Ingi Hong
- International Vaccine Institute, Gwanak-gu, Seoul, Korea
| | - Jung Hyuk Lee
- International Vaccine Institute, Gwanak-gu, Seoul, Korea
| | - Taewoo Kim
- International Vaccine Institute, Gwanak-gu, Seoul, Korea
| | - Sang Hwan Seo
- International Vaccine Institute, Gwanak-gu, Seoul, Korea
| | - Jae-Ouk Kim
- International Vaccine Institute, Gwanak-gu, Seoul, Korea
| | - Manki Song
- International Vaccine Institute, Gwanak-gu, Seoul, Korea
| | - Chung-Jong Kim
- Department of Internal Medicine, Ewha Womans University, Seoul, Korea
| | - Jae-Ki Choi
- Department of Infectious Diseases, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jieun Kim
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Eun Ju Choo
- Department of Infectious Diseases, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Jung-Hyun Choi
- Department of Infectious Diseases, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
53
|
Verheul MK, Vos M, de Rond L, De Zeeuw-Brouwer ML, Nijhof KH, Smit D, Oomen D, Molenaar P, Bogaard M, van Bergen R, Middelhof I, Beckers L, Wijmenga-Monsuur AJ, Buisman AM, Boer MC, van Binnendijk R, de Wit J, Guichelaar T. Contribution of SARS-CoV-2 infection preceding COVID-19 mRNA vaccination to generation of cellular and humoral immune responses in children. Front Immunol 2023; 14:1327875. [PMID: 38193077 PMCID: PMC10773747 DOI: 10.3389/fimmu.2023.1327875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Primary COVID-19 vaccination for children, 5-17 years of age, was offered in the Netherlands at a time when a substantial part of this population had already experienced a SARS-CoV-2 infection. While vaccination has been shown effective, underlying immune responses have not been extensively studied. We studied immune responsiveness to one and/or two doses of primary BNT162b2 mRNA vaccination and compared the humoral and cellular immune response in children with and without a preceding infection. Antibodies targeting the original SARS-CoV-2 Spike or Omicron Spike were measured by multiplex immunoassay. B-cell and T-cell responses were investigated using enzyme-linked immunosorbent spot (ELISpot) assays. The activation of CD4+ and CD8+ T cells was studied by flowcytometry. Primary vaccination induced both a humoral and cellular adaptive response in naive children. These responses were stronger in those with a history of infection prior to vaccination. A second vaccine dose did not further boost antibody levels in those who previously experienced an infection. Infection-induced responsiveness prior to vaccination was mainly detected in CD8+ T cells, while vaccine-induced T-cell responses were mostly by CD4+ T cells. Thus, SARS-CoV-2 infection prior to vaccination enhances adaptive cellular and humoral immune responses to primary COVID-19 vaccination in children. As most children are now expected to contract infection before the age of five, the impact of infection-induced immunity in children is of high relevance. Therefore, considering natural infection as a priming immunogen that enhances subsequent vaccine-responsiveness may help decision-making on the number and timing of vaccine doses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Teun Guichelaar
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
54
|
Ravlić S, Kurtović T, Cvetko Krajinović L, Hećimović A, Miloš M, Mateljak Lukačević S, Markotić A, Halassy B. What can neutralizing antibodies tell us about the quality of immunity in COVID-19 convalescents and vaccinees? Hum Vaccin Immunother 2023; 19:2270310. [PMID: 37905722 PMCID: PMC10760325 DOI: 10.1080/21645515.2023.2270310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/10/2023] [Indexed: 11/02/2023] Open
Abstract
During the SARS-CoV-2 pandemic, the lack of standardized measurements of the immune response after vaccination or recovery from COVID-19 resulted in incomparable results and hindered correlation establishment. Prioritizing reliable and standardized methods to monitor pathogen-specific immunity is crucial, not only during the COVID-19 pandemic but also for future outbreaks. During our study of the humoral immune response, we used a SARS-CoV-2 wild-type neutralization assay, ensuring the measurement of the immune response directed to all SARS-CoV-2 antigens in their proper conformation. A head-to-head comparison of the neutralizing antibody (NAb) responses elicited by four vaccines used in Europe during 2021 (BNT162b2, mRNA-1273, ChAdOx nCoV-19, and Ad26.COV2.S) and their comparison to NAb responses in convalescents showed that while the amount was comparable, NAbs induced by natural infection were of higher quality. Namely, NAbs produced by disease were better activators of the complement system than NAbs induced by vaccination. Furthermore, the contribution of spike protein-specific IgGs to the SARS-CoV-2 neutralization was lower in convalescents compared to vaccinees, indicating that those who recovered from COVID-19 were armed with antibodies of additional specificities and/or classes that contributed to virus neutralization. These findings suggest that a higher stringency of public policy measures targeting individuals who have recovered from COVID-19, in comparison to those who have been vaccinated, may not have been fully justified.
Collapse
Affiliation(s)
- Sanda Ravlić
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Virus Immunology and Vaccines, CERVirVac, Zagreb, Croatia
| | - Tihana Kurtović
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Virus Immunology and Vaccines, CERVirVac, Zagreb, Croatia
| | - Lidija Cvetko Krajinović
- Center of Excellence for Virus Immunology and Vaccines, CERVirVac, Zagreb, Croatia
- Research Department, University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, Zagreb, Croatia
| | - Ana Hećimović
- Service for Transfusion Medicine, Croatian Institute of Transfusion Medicine, Zagreb, Croatia
| | - Marija Miloš
- Clinical Department of Laboratory Diagnostic, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Sanja Mateljak Lukačević
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Virus Immunology and Vaccines, CERVirVac, Zagreb, Croatia
| | - Alemka Markotić
- Center of Excellence for Virus Immunology and Vaccines, CERVirVac, Zagreb, Croatia
- Research Department, University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, Zagreb, Croatia
| | - Beata Halassy
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Virus Immunology and Vaccines, CERVirVac, Zagreb, Croatia
| |
Collapse
|
55
|
Vellas C, Dimeglio C, Joncour E, Staes L, Jamme T, Miedougé M, Da-Silva I, Porcheron M, Migueres M, Kamar N, Izopet J. Evaluation of two anti-SARS-CoV-2 antibody immunoassays for monitoring patients on pre-exposure prophylaxis. Diagn Microbiol Infect Dis 2023; 107:116071. [PMID: 37716217 DOI: 10.1016/j.diagmicrobio.2023.116071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/31/2023] [Accepted: 08/22/2023] [Indexed: 09/18/2023]
Abstract
Pre-exposure prophylaxis (PrEP) is crucial to prevent severe COVID-19 in immunocompromised patients. A reliable method is needed to quantify anti-SARS-CoV-2 antibody levels for personalized monitoring during PrEP. We measured the binding antibody concentrations of 63 immunocompromised patients receiving 300mg or 600mg tixagevimab/cilgavimab on PrEP day and twice during the following 3 months. All blood samples were tested using the Abbott anti-SARS-CoV-2 IgG II Quant assay, the Roche Elecsys anti-SARS-CoV-2 S assay, and live virus-based neutralization assays. The results of the two immunoassays were correlated on day 0, 1 month, and 3 months post-PrEP. Passing-Bablok regression demonstrated higher anti-S concentration values measured with the Roche immunoassay compared to those measured with the Abbott immunoassay. Antibody concentrations were higher after 600 mg tixagevimab/cilgavimab prophylaxis than after 300 mg. The neutralizing antibody titers obtained using the omicron BA.5 and BA.2.75 strains were low. Both automated immunoassays are suitable for monitoring immunocompromised patients on PrEP.
Collapse
Affiliation(s)
- Camille Vellas
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France.
| | - Chloé Dimeglio
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse, France; CHU de Toulouse, Laboratoire de Virologie, Toulouse, France
| | - Emma Joncour
- CHU de Toulouse, Laboratoire de Virologie, Toulouse, France
| | - Laetitia Staes
- CHU de Toulouse, Laboratoire de Virologie, Toulouse, France
| | - Thibaut Jamme
- CHU de Toulouse, Laboratorie de Biochimie, Toulouse, France
| | | | | | | | - Marion Migueres
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; CHU de Toulouse, Laboratoire de Virologie, Toulouse, France
| | - Nassim Kamar
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; CHU de Toulouse, Département de Néphrologie, Dialyse et Transplantation d'Organes, Toulouse, France
| | - Jacques Izopet
- INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; CHU de Toulouse, Laboratoire de Virologie, Toulouse, France
| |
Collapse
|
56
|
Berry AA, Tjaden AH, Renteria J, Friedman-Klabanoff D, Hinkelman AN, Gibbs MA, Ahmed A, Runyon MS, Schieffelin J, Santos RP, Oberhelman R, Bott M, Correa A, Edelstein SL, Uschner D, Wierzba TF. Persistence of antibody responses to COVID-19 vaccines among participants in the COVID-19 Community Research Partnership. Vaccine X 2023; 15:100371. [PMID: 37649617 PMCID: PMC10462856 DOI: 10.1016/j.jvacx.2023.100371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 09/01/2023] Open
Abstract
Introduction High levels of immunity to SARS-CoV-2 in the community correlate with protection from COVID-19 illness. Measuring COVID-19 antibody seroprevalence and persistence may elucidate the level and length of protection afforded by vaccination and infection within a population. Methods We measured the duration of detectable anti-spike antibodies following COVID-19 vaccination in a multistate, longitudinal cohort study of almost 13,000 adults who completed daily surveys and submitted monthly dried blood spots collected at home. Results Overall, anti-spike antibodies persisted up to 284 days of follow-up with seroreversion occurring in only 2.4% of the study population. In adjusted analyses, risk of seroreversion increased with age (adults aged 55-64: adjusted hazard ratio [aHR] 2.19 [95% confidence interval (CI): 1.22, 3.92] and adults aged > 65: aHR 3.59 [95% CI: 2.07, 6.20] compared to adults aged 18-39). Adults with diabetes had a higher risk of seroreversion versus nondiabetics (aHR 1.77 [95% CI: 1.29, 2.44]). Decreased risk of seroreversion was shown for non-Hispanic Black versus non-Hispanic White (aHR 0.32 [95% CI: 0.13, 0.79]); college degree earners versus no college degree (aHR 0.61 [95% CI: 0.46, 0.81]); and those who received Moderna mRNA-1273 vaccine versus Pfizer-BioNTech BNT162b2 (aHR 0.35 [95% CI: 0.26, 0.47]). An interaction between healthcare worker occupation and sex was detected, with seroreversion increased among male, non-healthcare workers. Conclusion We established that a remote, longitudinal, multi-site study can reliably detect antibody durability following COVID-19 vaccination. The survey platform and measurement of antibody response using at-home collection at convenient intervals allowed us to explore sociodemographic factors and comorbidities and identify predictors of antibody persistence, which has been demonstrated to correlate with protection against disease. Our findings may help inform public health interventions and policies to protect those at highest risk for severe illness and assist in determining the optimal timing of booster doses.Clinical trials registry: NCT04342884.
Collapse
Affiliation(s)
- Andrea A. Berry
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ashley H. Tjaden
- The Biostatistics Center, Milken Institute School of Public Health, George Washington University, Rockville, MD, USA
| | - Jone Renteria
- The Biostatistics Center, Milken Institute School of Public Health, George Washington University, Rockville, MD, USA
| | - DeAnna Friedman-Klabanoff
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Amy N. Hinkelman
- Jerry M. Wallace School of Osteopathic Medicine, Campbell University, Lillington, NC, USA
| | | | | | | | - John Schieffelin
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Richard Oberhelman
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, USA
| | - Matthew Bott
- The Biostatistics Center, Milken Institute School of Public Health, George Washington University, Rockville, MD, USA
| | - Adolfo Correa
- University of Mississippi Medical Center, Jackson, MS, USA
| | - Sharon L. Edelstein
- The Biostatistics Center, Milken Institute School of Public Health, George Washington University, Rockville, MD, USA
| | - Diane Uschner
- The Biostatistics Center, Milken Institute School of Public Health, George Washington University, Rockville, MD, USA
| | - Thomas F. Wierzba
- Section on Infectious Diseases, Department of Internal Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | | |
Collapse
|
57
|
Flor N, García MI, Molineri A, Bottasso O, Diez C, Veaute C. Antibodies to SARS-CoV2 induced by vaccination and infection correlate with protection against the infection. Vaccine 2023; 41:7206-7211. [PMID: 37884413 DOI: 10.1016/j.vaccine.2023.10.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/03/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023]
Abstract
The COVID-19 pandemic remained worldwide for almost three years, but little is known about the dynamics of humoral immune response to the third dose over time and its protection from infection. Our aim was to assess the humoral immune response after the third dose of the different vaccines administered to SARS-CoV-2 naive and previously infected individuals, and its correlation with protection in an academic community. For each person studied (185), three blood samples were taken between December 2021 and July 2022, one month apart. Anti-S antibodies were quantified by ELISA, while anti-N antibody levels were determined by ECLIA. Most of the participants had received two doses of viral vector-based, mRNA-based and virus-inactivated vaccines. Although anti-N antibody levels revealed that 80% of the individuals had been exposed to the virus before or during the study, only 42% reported having been diagnosed. When anti-S IgG levels were measured 3-5 months after the second dose of any vaccine, they were higher in those previously infected individuals. The same results were observed for anti-N IgG levels in those who received 2 doses of the virus-inactivated vaccine. When analyzing the dynamics of anti-S antibodies we observed that, although positive IgG antibody levels were detected 5-6 months after the second dose administration, those observed 30-60 days after the third dose were significantly higher and remained so for at least 8 months. Higher levels of anti-S IgG antibodies at the first sampling were associated with a lower incidence of subsequent infection. The same association was seen in people who received the booster compared with those who received two doses. This study provides further evidence that anti-S IgG antibodies remained at high levels over time, and both anti-S levels and the third dose of anti-SARS-CoV-2 vaccine correlate with protection against the infection. It also shows that infection acts as a booster of immunization, increasing levels of both anti-N and anti-S IgG.
Collapse
Affiliation(s)
- Noelia Flor
- Laboratorio de Inmunología Experimental, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Argentina
| | - María Inés García
- Laboratorio de Inmunología Experimental, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Argentina
| | - Ana Molineri
- Instituto de Investigación de la Cadena Láctea, Instituto Nacional de Tecnología Agropecuaria-CONICET, Argentina
| | - Oscar Bottasso
- Instituto de Inmunología Clínica y Experimental de Rosario, Universidad Nacional de Rosario. CONICET, Argentina
| | - Cristina Diez
- Laboratorio de Biología Molecular e Inmunología Aplicadas, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Argentina
| | - Carolina Veaute
- Laboratorio de Inmunología Experimental, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Argentina; Laboratorio de Biología Molecular e Inmunología Aplicadas, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Argentina.
| |
Collapse
|
58
|
Okuyama R. mRNA and Adenoviral Vector Vaccine Platforms Utilized in COVID-19 Vaccines: Technologies, Ecosystem, and Future Directions. Vaccines (Basel) 2023; 11:1737. [PMID: 38140142 PMCID: PMC10748114 DOI: 10.3390/vaccines11121737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/12/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
New technological platforms, such as mRNA and adenoviral vector vaccines, have been utilized to develop coronavirus disease 2019 (COVID-19) vaccines. These new modalities enable rapid and flexible vaccine design and cost-effective and swift manufacturing, effectively combating pandemics caused by mutating viruses. Innovation ecosystems, including universities, startups, investors, and governments are crucial for developing these cutting-edge technologies. This review summarizes the research and development trajectory of these vaccine technologies, their investments, and the support surrounding them, in addition to the technological details of each technology. In addition, this study examines the importance of an innovation ecosystem in developing novel technologies, comparing it with the case of Japan, which has lagged behind in COVID-19 vaccine development. It also explores the direction of vaccine development in the post-COVID-19 era.
Collapse
Affiliation(s)
- Ryo Okuyama
- College of International Management, Ritsumeikan Asia Pacific University, Beppu 874-8577, Japan
| |
Collapse
|
59
|
Hvidt AK, Guo H, Andersen R, Lende SSF, Vibholm LK, Søgaard OS, Schleimann MH, Russell V, Cheung AMW, Paramithiotis E, Olesen R, Tolstrup M. Long-term humoral and cellular immunity after primary SARS-CoV-2 infection: a 20-month longitudinal study. BMC Immunol 2023; 24:45. [PMID: 37974069 PMCID: PMC10652616 DOI: 10.1186/s12865-023-00583-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND SARS-CoV-2 remains a world-wide health issue. SARS-CoV-2-specific immunity is induced upon both infection and vaccination. However, defining the long-term immune trajectory, especially after infection, is limited. In this study, we aimed to further the understanding of long-term SARS-CoV-2-specific immune response after infection. RESULTS We conducted a longitudinal cohort study among 93 SARS-CoV-2 recovered individuals. Immune responses were continuously monitored for up to 20 months after infection. The humoral responses were quantified by Spike- and Nucleocapsid-specific IgG levels. T cell responses to Spike- and non-Spike epitopes were examined using both intercellular cytokine staining (ICS) assay and Activation-Induced marker (AIM) assay with quantification of antigen-specific IFNγ production. During the 20 months follow-up period, Nucleocapsid-specific antibody levels and non-Spike-specific CD4 + and CD8 + T cell frequencies decreased in the blood. However, a majority of participants maintained a durable immune responses 20 months after infection: 59% of the participants were seropositive for Nucleocapsid-specific IgG, and more than 70% had persisting non-Spike-specific T cells. The Spike-specific response initially decreased but as participants were vaccinated against COVID-19, Spike-specific IgG levels and T cell frequencies were boosted reaching similar or higher levels compared to 1 month post-infection. The trajectory of infection-induced SARS-CoV-2-specific immunity decreases, but for the majority of participants it persists beyond 20 months. The T cell response displays a greater durability. Vaccination boosts Spike-specific immune responses to similar or higher levels as seen after primary infection. CONCLUSIONS For most participants, the response persists 20 months after infection, and the cellular response appears to be more long-lived compared to the circulating antibody levels. Vaccination boosts the S-specific response but does not affect the non-S-specific response. Together, these findings support the understanding of immune contraction, and with studies showing the immune levels required for protection, adds to the knowledge of durability of protection against future SARS-CoV-2.
Collapse
Affiliation(s)
- Astrid Korning Hvidt
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Rebecca Andersen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Stine Sofie Frank Lende
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Line Khalidan Vibholm
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ole Schmeltz Søgaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Marianne Hoegsbjerg Schleimann
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Victoria Russell
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Angela Man-Wei Cheung
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Medicine, University Health Network, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | | | - Rikke Olesen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark.
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
60
|
Yap DYH, Fong CHY, Zhang X, Ip JD, Chan WM, Chu AWH, Chen LL, Zhao Y, Chan BPC, Luk KS, Cheng VCC, Chan TM, To KKW. Humoral and cellular immunity against different SARS-CoV-2 variants in patients with chronic kidney disease. Sci Rep 2023; 13:19932. [PMID: 37968273 PMCID: PMC10652016 DOI: 10.1038/s41598-023-47130-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/09/2023] [Indexed: 11/17/2023] Open
Abstract
Chronic kidney disease (CKD) patients are at higher risk of severe COVID-19. Humoral and cellular immunity from prior infection or vaccination are important for protection, but the neutralizing antibody (nAb) response against SARS-CoV-2 variants is impaired. We investigated the variant-specific nAb and T cell immunity among CKD patients. Adult CKD patients were recruited between August and October 2022. nAb against the SARS-CoV-2 (ancestral strains and four Omicron sublineages) and T cell response were measured using the live virus neutralization assay and interferon-gamma release assay (IGRA). The correlation between nAb/T-cell response and subsequent infection after recruitment were also determined. Among the 88 recruited patients, 95.5% had prior infection or had completed the primary vaccine series. However, only 77.3% had detectable nAb against at least one SARS-CoV-2 strains, 59.1% tested positive in IGRA, and 52.3% had detectable nAb and tested positive in the IGRA. The nAb geometic mean titers (GMTs) against XBB.1, BA.5 and BA.2.3.20 were significantly lower than those against BA.2 and ancestral strain. Prior SARS-CoV-2 infection was associated with elevated nAb and T cell response. More kidney transplant recipients (KTRs) showed absent nAb and T cell response (36.8% vs. 10.1%), despite a higher prevalence of vaccine booster in this population (94.7% vs. 50.7%). Lower levels of nAb titer and T cell response were significantly associated with subsequent infection. A considerable proportion of CKD patients, especially KTRs, showed absence of humoral and cellular protective immunity against SARS-CoV-2. Strategies to improve immunogenicity in this population are urgently needed.
Collapse
Affiliation(s)
- Desmond Yat-Hin Yap
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Carol Ho-Yan Fong
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, People's Republic of China
| | - Xiaojuan Zhang
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Jonathan Daniel Ip
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Wan-Mui Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Allen Wing-Ho Chu
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, People's Republic of China
| | - Lin-Lei Chen
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Yan Zhao
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Brian Pui-Chun Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Kristine Shik Luk
- Department of Pathology, Princess Margaret Hospital, Kwai Chung, Hong Kong Special Administrative Region, People's Republic of China
| | - Vincent Chi-Chung Cheng
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Tak-Mao Chan
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Kelvin Kai-Wang To
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Shatin, Hong Kong Special Administrative Region, People's Republic of China.
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People's Republic of China.
| |
Collapse
|
61
|
Polonsky K, Pupko T, Freund NT. Evaluation of the Ability of AlphaFold to Predict the Three-Dimensional Structures of Antibodies and Epitopes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1578-1588. [PMID: 37782047 DOI: 10.4049/jimmunol.2300150] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/06/2023] [Indexed: 10/03/2023]
Abstract
Being able to accurately predict the three-dimensional structure of an Ab can facilitate Ab characterization and epitope prediction, with important diagnostic and clinical implications. In this study, we evaluated the ability of AlphaFold to predict the structures of 222 recently published, high-resolution Fab H and L chain structures of Abs from different species directed against different Ags. We show that although the overall Ab prediction quality is in line with the results of CASP14, regions such as the complementarity-determining regions (CDRs) of the H chain, which are prone to higher variation, are predicted less accurately. Moreover, we discovered that AlphaFold mispredicts the bending angles between the variable and constant domains. To evaluate the ability of AlphaFold to model Ab-Ag interactions based only on sequence, we used AlphaFold-Multimer in combination with ZDOCK to predict the structures of 26 known Ab-Ag complexes. ZDOCK, which was applied on bound components of both the Ab and the Ag, succeeded in assembling 11 complexes, whereas AlphaFold succeeded in predicting only 2 of 26 models, with significant deviations in the docking contacts predicted in the rest of the molecules. Within the 11 complexes that were successfully predicted by ZDOCK, 9 involved short-peptide Ags (18-mer or less), whereas only 2 were complexes of Ab with a full-length protein. Docking of modeled unbound Ab and Ag was unsuccessful. In summary, our study provides important information about the abilities and limitations of using AlphaFold to predict Ab-Ag interactions and suggests areas for possible improvement.
Collapse
Affiliation(s)
- Ksenia Polonsky
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tal Pupko
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Natalia T Freund
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
62
|
Yoon JG, Kim YE, Choi MJ, Choi WS, Seo YB, Jung J, Hyun HJ, Seong H, Nham E, Noh JY, Song JY, Kim WJ, Kim DW, Cheong HJ. Herpes Zoster Reactivation After mRNA and Adenovirus-Vectored Coronavirus Disease 2019 Vaccination: Analysis of National Health Insurance Database. J Infect Dis 2023; 228:1326-1335. [PMID: 37549237 PMCID: PMC10640769 DOI: 10.1093/infdis/jiad297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/20/2023] [Accepted: 07/26/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Our study aimed to determine the risk of herpes zoster reactivation and coronavirus disease 2019 (COVID-19) vaccination (mRNA vaccine [BNT162b2] and adenovirus-vectored vaccine [ChAdOx1 nCoV-19]). METHODS This retrospective study analyzed herpes zoster cases diagnosed between 26 February 2021 and 30 June 2021 and registered in the National Health Insurance Service database. A matched case-control study with a 1:3 matching ratio and a propensity score matching (PSM) study with a 1:1 ratio of vaccinated and unvaccinated individuals were performed. RESULTS In the matched case control analysis, BNT162b2 was associated with an increased risk of herpes zoster reactivation (first dose adjusted odds ratio [aOR], 1.11; 95% confidence interval [CI], 1.06-1.15; second dose aOR, 1.17; 95% CI, 1.12-1.23). PSM analysis revealed a statistically significant increase in risk within 18 days following any vaccination (adjusted hazard ratio [aHR], 1.09; 95% CI, 1.02-1.16). BNT162b2 was associated with an increased risk at 18 days postvaccination (aHR, 1.65; 95% CI, 1.35-2.02) and second dose (aHR, 1.10; 95% CI, 1.02-1.19). However, the risk did not increase in both analyses of ChAdOx1 vaccination. CONCLUSIONS mRNA COVID-19 vaccination possibly increases the risk of herpes zoster reactivation, and thus close follow-up for herpes zoster reactivation is required.
Collapse
Affiliation(s)
- Jin Gu Yoon
- Division of Infectious Diseases, Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Young-Eun Kim
- Big Data Department, National Health Insurance Service, Wonju, South Korea
| | - Min Joo Choi
- Department of Internal Medicine, International St Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon, South Korea
| | - Won Suk Choi
- Division of Infectious Diseases, Department of Internal Medicine, Ansan Hospital, Korea University College of Medicine, Ansan, South Korea
| | - Yu Bin Seo
- Division of Infectious Diseases, Department of Internal Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea
| | - Jaehun Jung
- Artificial Intelligence and Big-Data Convergence Center, Gachon University College of Medicine, Incheon, South Korea
| | - Hak-Jun Hyun
- Division of Infectious Diseases, Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Hye Seong
- Division of Infectious Diseases, Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Eliel Nham
- Division of Infectious Diseases, Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Ji Yun Noh
- Division of Infectious Diseases, Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Joon Young Song
- Division of Infectious Diseases, Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Woo Joo Kim
- Division of Infectious Diseases, Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Dong Wook Kim
- Department of Information and Statistics, Department of Bio and Medical Big Data, Research Institute of Natural Science, Gyeongsang National University, Jinju, South Korea
| | - Hee Jin Cheong
- Division of Infectious Diseases, Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| |
Collapse
|
63
|
Neale I, Ali M, Kronsteiner B, Longet S, Abraham P, Deeks AS, Brown A, Moore SC, Stafford L, Dobson SL, Plowright M, Newman TAH, Wu MY, Carr EJ, Beale R, Otter AD, Hopkins S, Hall V, Tomic A, Payne RP, Barnes E, Richter A, Duncan CJA, Turtle L, de Silva TI, Carroll M, Lambe T, Klenerman P, Dunachie S. 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 2023; 14:e0121223. [PMID: 37655880 PMCID: PMC10653804 DOI: 10.1128/mbio.01212-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/26/2023] [Indexed: 09/02/2023] Open
Abstract
IMPORTANCE Defining correlates of protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine breakthrough infection informs vaccine policy for booster doses and future vaccine designs. Existing studies demonstrate humoral correlates of protection, but the role of T cells in protection is still unclear. In this study, we explore antibody and T cell immune responses associated with protection against Delta variant vaccine breakthrough infection in a well-characterized cohort of UK Healthcare Workers (HCWs). We demonstrate evidence to support a role for CD4+ and CD8+ T cells as well as antibodies against Delta vaccine breakthrough infection. In addition, our results suggest a potential role for cross-reactive T cells in vaccine breakthrough.
Collapse
Affiliation(s)
- Isabel Neale
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| | - Mohammad Ali
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Stephanie Longet
- Nuffield Department of Medicine, Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Priyanka Abraham
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Alexandra S. Deeks
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Anthony Brown
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Shona C. Moore
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Lizzie Stafford
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Susan L. Dobson
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Megan Plowright
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Thomas A. H. Newman
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Mary Y. Wu
- Covid Surveillance Unit, The Francis Crick Institute, London, United Kingdom
| | - Crick COVID Immunity Pipeline
- Covid Surveillance Unit, The Francis Crick Institute, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | | | - Rupert Beale
- The Francis Crick Institute, London, United Kingdom
- UCL Department of Renal Medicine, Royal Free Hospital, London, United Kingdom
| | | | | | | | - Adriana Tomic
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Rebecca P. Payne
- Translational and Clinical Research Institute Immunity and Inflammation Theme, Newcastle University, Newcastle, United Kingdom
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Alex Richter
- Institute of Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Christopher J. A. Duncan
- Translational and Clinical Research Institute Immunity and Inflammation Theme, Newcastle University, Newcastle, United Kingdom
- Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | - Lance Turtle
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Thushan I. de Silva
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Miles Carroll
- Nuffield Department of Medicine, Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Teresa Lambe
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Susanna Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - On behalf of the PITCH Consortium
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
- Nuffield Department of Medicine, Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
- Covid Surveillance Unit, The Francis Crick Institute, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
- UCL Department of Renal Medicine, Royal Free Hospital, London, United Kingdom
- UK Health Security Agency, Porton Down, United Kingdom
- UK Health Security Agency, London, United Kingdom
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
- Translational and Clinical Research Institute Immunity and Inflammation Theme, Newcastle University, Newcastle, United Kingdom
- Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Institute of Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, United Kingdom
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| |
Collapse
|
64
|
Wang L, Patrone PN, Kearsley AJ, Izac JR, Gaigalas AK, Prostko JC, Kwon HJ, Tang W, Kosikova M, Xie H, Tian L, Elsheikh EB, Kwee EJ, Kemp T, Jochum S, Thornburg N, McDonald LC, Gundlapalli AV, Lin-Gibson S. Monoclonal Antibodies as SARS-CoV-2 Serology Standards: Experimental Validation and Broader Implications for Correlates of Protection. Int J Mol Sci 2023; 24:15705. [PMID: 37958688 PMCID: PMC10650176 DOI: 10.3390/ijms242115705] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
COVID-19 has highlighted challenges in the measurement quality and comparability of serological binding and neutralization assays. Due to many different assay formats and reagents, these measurements are known to be highly variable with large uncertainties. The development of the WHO international standard (WHO IS) and other pool standards have facilitated assay comparability through normalization to a common material but does not provide assay harmonization nor uncertainty quantification. In this paper, we present the results from an interlaboratory study that led to the development of (1) a novel hierarchy of data analyses based on the thermodynamics of antibody binding and (2) a modeling framework that quantifies the probability of neutralization potential for a given binding measurement. Importantly, we introduced a precise, mathematical definition of harmonization that separates the sources of quantitative uncertainties, some of which can be corrected to enable, for the first time, assay comparability. Both the theory and experimental data confirmed that mAbs and WHO IS performed identically as a primary standard for establishing traceability and bridging across different assay platforms. The metrological anchoring of complex serological binding and neuralization assays and fast turn-around production of an mAb reference control can enable the unprecedented comparability and traceability of serological binding assay results for new variants of SARS-CoV-2 and immune responses to other viruses.
Collapse
Affiliation(s)
- Lili Wang
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (J.R.I.); (A.K.G.); (L.T.); (E.B.E.); (E.J.K.)
| | - Paul N. Patrone
- Applied and Computational Mathematics Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (P.N.P.); (A.J.K.)
| | - Anthony J. Kearsley
- Applied and Computational Mathematics Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (P.N.P.); (A.J.K.)
| | - Jerilyn R. Izac
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (J.R.I.); (A.K.G.); (L.T.); (E.B.E.); (E.J.K.)
| | - Adolfas K. Gaigalas
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (J.R.I.); (A.K.G.); (L.T.); (E.B.E.); (E.J.K.)
| | | | - Hyung Joon Kwon
- Laboratory of Pediatric and Respiratory Viral Diseases, Office of Vaccines Research and Review, Center for Biologics Evaluation, Food and Drug Administration (FDA), Silver Spring, MD 20993, USA; (H.J.K.); (W.T.); (M.K.); (H.X.)
| | - Weichun Tang
- Laboratory of Pediatric and Respiratory Viral Diseases, Office of Vaccines Research and Review, Center for Biologics Evaluation, Food and Drug Administration (FDA), Silver Spring, MD 20993, USA; (H.J.K.); (W.T.); (M.K.); (H.X.)
| | - Martina Kosikova
- Laboratory of Pediatric and Respiratory Viral Diseases, Office of Vaccines Research and Review, Center for Biologics Evaluation, Food and Drug Administration (FDA), Silver Spring, MD 20993, USA; (H.J.K.); (W.T.); (M.K.); (H.X.)
| | - Hang Xie
- Laboratory of Pediatric and Respiratory Viral Diseases, Office of Vaccines Research and Review, Center for Biologics Evaluation, Food and Drug Administration (FDA), Silver Spring, MD 20993, USA; (H.J.K.); (W.T.); (M.K.); (H.X.)
| | - Linhua Tian
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (J.R.I.); (A.K.G.); (L.T.); (E.B.E.); (E.J.K.)
| | - Elzafir B. Elsheikh
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (J.R.I.); (A.K.G.); (L.T.); (E.B.E.); (E.J.K.)
| | - Edward J. Kwee
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (J.R.I.); (A.K.G.); (L.T.); (E.B.E.); (E.J.K.)
| | - Troy Kemp
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD 21702, USA;
| | - Simon Jochum
- Roche Diagnostics GmbH, 82377 Penzberg, Germany;
| | - Natalie Thornburg
- Centers for Disease Control and Prevention (CDC), Atlanta, GA 30329, USA; (N.T.); (L.C.M.); (A.V.G.)
| | - L. Clifford McDonald
- Centers for Disease Control and Prevention (CDC), Atlanta, GA 30329, USA; (N.T.); (L.C.M.); (A.V.G.)
| | - Adi V. Gundlapalli
- Centers for Disease Control and Prevention (CDC), Atlanta, GA 30329, USA; (N.T.); (L.C.M.); (A.V.G.)
| | - Sheng Lin-Gibson
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (J.R.I.); (A.K.G.); (L.T.); (E.B.E.); (E.J.K.)
| |
Collapse
|
65
|
Jay C, Adland E, Csala A, Lim N, Longet S, Ogbe A, Ratcliff J, Sampson O, Thompson CP, Turtle L, Barnes E, Dunachie S, Klenerman P, Carroll M, Goulder P. Age- and sex-specific differences in immune responses to BNT162b2 COVID-19 and live-attenuated influenza vaccines in UK adolescents. Front Immunol 2023; 14:1248630. [PMID: 37942333 PMCID: PMC10627794 DOI: 10.3389/fimmu.2023.1248630] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/15/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction The key to understanding the COVID-19 correlates of protection is assessing vaccine-induced immunity in different demographic groups. Young people are at a lower risk of COVID-19 mortality, females are at a lower risk than males, and females often generate stronger immune responses to vaccination. Methods We studied immune responses to two doses of BNT162b2 Pfizer COVID-19 vaccine in an adolescent cohort (n = 34, ages 12-16), an age group previously shown to elicit significantly greater immune responses to the same vaccine than young adults. Adolescents were studied with the aim of comparing their response to BNT162b2 to that of adults; and to assess the impacts of other factors such as sex, ongoing SARS-CoV-2 infection in schools, and prior exposure to endemic coronaviruses that circulate at high levels in young people. At the same time, we were able to evaluate immune responses to the co-administered live attenuated influenza vaccine. Blood samples from 34 adolescents taken before and after vaccination with COVID-19 and influenza vaccines were assayed for SARS-CoV-2-specific IgG and neutralising antibodies and cellular immunity specific for SARS-CoV-2 and endemic betacoronaviruses. The IgG targeting influenza lineages contained in the influenza vaccine were also assessed. Results Robust neutralising responses were identified in previously infected adolescents after one dose, and two doses were required in infection-naïve adolescents. As previously demonstrated, total IgG responses to SARS-CoV-2 Spike were significantly higher among vaccinated adolescents than among adults (aged 32-52) who received the BNT162b2 vaccine (comparing infection-naïve, 49,696 vs. 33,339; p = 0.03; comparing SARS-CoV-2 previously infected, 743,691 vs. 269,985; p <0.0001) by the MSD v-plex assay. There was no evidence of a stronger vaccine-induced immunity in females compared than in males. Discussion These findings may result from the introduction of novel mRNA vaccination platforms, generating patterns of immunity divergent from established trends and providing new insights into what might be protective following COVID-19 vaccination.
Collapse
Affiliation(s)
- Cecilia Jay
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Emily Adland
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Anna Csala
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Nicholas Lim
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Stephanie Longet
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Ane Ogbe
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jeremy Ratcliff
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Oliver Sampson
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Craig P. Thompson
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Warwick, United Kingdom
| | - Lance Turtle
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Eleanor Barnes
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Susanna Dunachie
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Paul Klenerman
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Miles Carroll
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Philip Goulder
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
66
|
Fryer HA, Hartley GE, Edwards ESJ, Varese N, Boo I, Bornheimer SJ, Hogarth PM, Drummer HE, O'Hehir RE, van Zelm MC. COVID-19 Adenoviral Vector Vaccination Elicits a Robust Memory B Cell Response with the Capacity to Recognize Omicron BA.2 and BA.5 Variants. J Clin Immunol 2023; 43:1506-1518. [PMID: 37322095 PMCID: PMC10499924 DOI: 10.1007/s10875-023-01527-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/27/2023] [Indexed: 06/17/2023]
Abstract
Following the COVID-19 pandemic, novel vaccines have successfully reduced severe disease and death. Despite eliciting lower antibody responses, adenoviral vector vaccines are nearly as effective as mRNA vaccines. Therefore, protection against severe disease may be mediated by immune memory cells. We here evaluated plasma antibody and memory B cells (Bmem) targeting the SARS-CoV-2 Spike receptor-binding domain (RBD) elicited by the adenoviral vector vaccine ChAdOx1 (AstraZeneca), their capacity to bind Omicron subvariants, and compared this to the response to mRNA BNT162b2 (Pfizer-BioNTech) vaccination. Whole blood was sampled from 31 healthy adults pre-vaccination and 4 weeks after dose one and dose two of ChAdOx1. Neutralizing antibodies (NAb) against SARS-CoV-2 were quantified at each time point. Recombinant RBDs of the Wuhan-Hu-1 (WH1), Delta, BA.2, and BA.5 variants were produced for ELISA-based quantification of plasma IgG and incorporated separately into fluorescent tetramers for flow cytometric identification of RBD-specific Bmem. NAb and RBD-specific IgG levels were over eight times lower following ChAdOx1 vaccination than BNT162b2. In ChAdOx1-vaccinated individuals, median plasma IgG recognition of BA.2 and BA.5 as a proportion of WH1-specific IgG was 26% and 17%, respectively. All donors generated resting RBD-specific Bmem, which were boosted after the second dose of ChAdOx1 and were similar in number to those produced by BNT162b2. The second dose of ChAdOx1 boosted Bmem that recognized VoC, and 37% and 39% of WH1-specific Bmem recognized BA.2 and BA.5, respectively. These data uncover mechanisms by which ChAdOx1 elicits immune memory to confer effective protection against severe COVID-19.
Collapse
Affiliation(s)
- Holly A Fryer
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Gemma E Hartley
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Emily S J Edwards
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Nirupama Varese
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Immune Therapies Group, Burnet Institute, Melbourne, VIC, Australia
| | - Irene Boo
- Viral Entry and Vaccines Group, Burnet Institute, Melbourne, VIC, Australia
| | | | - P Mark Hogarth
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Immune Therapies Group, Burnet Institute, Melbourne, VIC, Australia
- Department of Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Heidi E Drummer
- Viral Entry and Vaccines Group, Burnet Institute, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Robyn E O'Hehir
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, VIC, Australia
| | - Menno C van Zelm
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia.
- Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, VIC, Australia.
| |
Collapse
|
67
|
Jarlhelt I, Pérez-Alós L, Bayarri-Olmos R, Hansen CB, Petersen MS, Weihe P, Armenteros JJA, Madsen JR, Nielsen JPS, Hilsted LM, Iversen KK, Bundgaard H, Nielsen SD, Garred P. Distinguishing SARS-CoV-2 infection and vaccine responses up to 18 months post-infection using nucleocapsid protein and receptor-binding domain antibodies. Microbiol Spectr 2023; 11:e0179623. [PMID: 37738355 PMCID: PMC10580960 DOI: 10.1128/spectrum.01796-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/04/2023] [Indexed: 09/24/2023] Open
Abstract
The prediction of the durability of immunity against COVID-19 is relevant, and longitudinal studies are essential for unraveling the details regarding protective SARS-CoV-2 antibody responses. It has become challenging to discriminate between COVID-19 vaccine- and infection-induced immune responses since all approved vaccines in Europe and the USA are based on the viral spike (S) protein, which is also the most commonly used antigen in immunoassays measuring immunoglobulins (Igs) against SARS-CoV-2. We have developed a nucleocapsid (N) protein-based sandwich ELISA for detecting pan anti-SARS-CoV-2 Ig with a sensitivity and specificity of 97%. Generalized mixed models were used to determine the degree of long-term humoral immunity against the N protein and the receptor-binding domain (RBD) of the S protein in a cohort of infected individuals to distinguish between COVID-19 vaccine- and infection-induced immunity. N-specific waning could be observed in individuals who did not experience reinfection, while individuals who experienced reinfection had a new significant increase in N-specific Ig levels. In individuals that seroconverted without a reinfection, 70.1% remained anti-N seropositive after 550 days. The anti-RBD Ig dynamics were unaffected by reinfection but exhibited a clear increase in RBD-specific Ig when vaccination was initiated. In conclusion, a clear difference in the dynamics of the antibody response against N protein and RBD was observed over time. Anti-N protein-specific Igs can be detected up to 18 months after SARS-CoV-2 infection allowing long-term discrimination of infectious and vaccine antibody responses.IMPORTANCELongitudinal studies are essential to unravel details regarding the protective antibody responses after COVID-19 infection and vaccination. It has become challenging to distinguish long-term immune responses to SARS-CoV-2 infection and vaccination since most approved vaccines are based on the viral spike (S) protein, which is also mostly used in immunoassays measuring immunoglobulins (Igs) against SARS-CoV-2. We have developed a novel nucleocapsid (N) protein-based sandwich ELISA for detecting pan-anti-SARS-CoV-2 Ig, exhibiting high sensitivity and specificity. Generalized mixed models were used to determine long-term humoral immunity in a cohort of infected individuals from the Faroe Islands, distinguishing between COVID-19 vaccine- and infection-induced immunity. A clear difference in the dynamics of the antibody response against N protein and S protein was observed over time, and the anti-N protein-specific Igs could be detected up to 18 months after SARS-CoV-2 infection. This enables long-term discrimination between natural infection and vaccine-dependent antibody responses.
Collapse
Affiliation(s)
- Ida Jarlhelt
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
| | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
| | - Rafael Bayarri-Olmos
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
- Recombinant Protein and Antibody Unit, Copenhagen University Hospital, Copenhagen, Denmark
| | - Cecilie Bo Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
| | - Maria Skaalum Petersen
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands, Denmark
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands, Denmark
| | - Pál Weihe
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands, Denmark
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands, Denmark
| | | | - Johannes Roth Madsen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
- Department of Emergency Medicine, Herlev-Gentofte Hospital, Copenhagen, Denmark
| | - Jacob Pohl Stangerup Nielsen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
- Department of Emergency Medicine, Herlev-Gentofte Hospital, Copenhagen, Denmark
| | | | - Kasper Karmark Iversen
- Department of Emergency Medicine, Herlev-Gentofte Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Heart Center, Department of Cardiology, Rigshospitalet, Copenhagen, Denmark
| | - Susanne Dam Nielsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Viro-immunology Research Unit, Department of Infectious Diseases, Section 8632, Rigshospitalet, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
68
|
Barreto MDS, da Silva BS, Santos RS, Silva DMRR, Silva EED, Moura PHM, de Souza JB, Santana LADM, Fonseca DLM, Filgueiras IS, Guimarães AG, Cabral-Marques O, Schimke LF, Borges LP. COVID-19 Vaccination and Serological Profile of a Brazilian University Population. Life (Basel) 2023; 13:1925. [PMID: 37763328 PMCID: PMC10532467 DOI: 10.3390/life13091925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND COVID-19 led to the suspension academic activities worldwide, affecting millions of students and staff. METHODS In this study, we evaluated the presence of IgM and IgG anti-SARS-CoV-2 antibodies in an academic population during the return to classes after a one-year suspension. The study took place over five months at a Brazilian university and included 942 participants. RESULTS We found that most participants had reactive IgG and non-reactive IgM. All received at least one dose, and 940 received two or more doses, of different COVID-19 vaccines. We obtained a higher average of memory antibodies (IgG) in participants who received the CoronaVac/ChAdOx1 combination. IgG was consistently distributed for each vaccine group, but individuals who completed the vaccination schedule had higher levels. There were no differences between antibodies and gender, presence of symptoms, and previous COVID-19 infection, but older participants (>53 years) and contacts of infected individuals had higher IgM levels. CONCLUSION This study makes significant contributions to the assessment of antibodies in the academic environment, allowing us to infer that most participants had memory immunity and low indications of recent infection when returning to face-to-face classes, as well as demonstrating the need to monitor immunity and update vaccinations.
Collapse
Affiliation(s)
- Marina dos Santos Barreto
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão 49100-000, SE, Brazil; (M.d.S.B.); (B.S.d.S.); (R.S.S.); (D.M.R.R.S.); (E.E.D.S.); (P.H.M.M.); (J.B.d.S.); (A.G.G.)
| | - Beatriz Soares da Silva
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão 49100-000, SE, Brazil; (M.d.S.B.); (B.S.d.S.); (R.S.S.); (D.M.R.R.S.); (E.E.D.S.); (P.H.M.M.); (J.B.d.S.); (A.G.G.)
| | - Ronaldy Santana Santos
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão 49100-000, SE, Brazil; (M.d.S.B.); (B.S.d.S.); (R.S.S.); (D.M.R.R.S.); (E.E.D.S.); (P.H.M.M.); (J.B.d.S.); (A.G.G.)
| | - Deise Maria Rego Rodrigues Silva
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão 49100-000, SE, Brazil; (M.d.S.B.); (B.S.d.S.); (R.S.S.); (D.M.R.R.S.); (E.E.D.S.); (P.H.M.M.); (J.B.d.S.); (A.G.G.)
| | - Eloia Emanuelly Dias Silva
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão 49100-000, SE, Brazil; (M.d.S.B.); (B.S.d.S.); (R.S.S.); (D.M.R.R.S.); (E.E.D.S.); (P.H.M.M.); (J.B.d.S.); (A.G.G.)
| | - Pedro Henrique Macedo Moura
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão 49100-000, SE, Brazil; (M.d.S.B.); (B.S.d.S.); (R.S.S.); (D.M.R.R.S.); (E.E.D.S.); (P.H.M.M.); (J.B.d.S.); (A.G.G.)
| | - Jessiane Bispo de Souza
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão 49100-000, SE, Brazil; (M.d.S.B.); (B.S.d.S.); (R.S.S.); (D.M.R.R.S.); (E.E.D.S.); (P.H.M.M.); (J.B.d.S.); (A.G.G.)
| | | | - Dennyson Leandro M. Fonseca
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of São Paulo (USP), São Paulo 05508-090, SP, Brazil;
| | - Igor Salerno Filgueiras
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil;
| | - Adriana Gibara Guimarães
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão 49100-000, SE, Brazil; (M.d.S.B.); (B.S.d.S.); (R.S.S.); (D.M.R.R.S.); (E.E.D.S.); (P.H.M.M.); (J.B.d.S.); (A.G.G.)
| | - Otavio Cabral-Marques
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of São Paulo (USP), São Paulo 05508-090, SP, Brazil;
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil;
- Department of Medicine, Division of Molecular Medicine, School of Medicine, University of São Paulo, São Paulo 01246-903, SP, Brazil
- Department of Pharmacy and Postgraduate Program of Health and Science, Federal University of Rio Grande do Norte, Natal 59012-570, RN, Brazil
- Laboratory of Medical Investigation 29, University of São Paulo School of Medicine, São Paulo 01246-903, SP, Brazil
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
- Network of Immunity in Infection, Malignancy, Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo 05508-000, SP, Brazil
| | - Lena F. Schimke
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil;
- Department of Medicine, Division of Molecular Medicine, School of Medicine, University of São Paulo, São Paulo 01246-903, SP, Brazil
| | - Lysandro Pinto Borges
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão 49100-000, SE, Brazil; (M.d.S.B.); (B.S.d.S.); (R.S.S.); (D.M.R.R.S.); (E.E.D.S.); (P.H.M.M.); (J.B.d.S.); (A.G.G.)
| |
Collapse
|
69
|
Baerends EA, Hvidt AK, Reekie J, Søgaard OS, Stærke NB, Raben D, Nielsen H, Petersen KT, Juhl MR, Johansen IS, Lindvig SO, Madsen LW, Wiese L, Knudsen LS, Iversen MB, Benfield T, Iversen KK, Andersen SD, Juhl AK, Dietz LL, Andreasen SR, Fischer TK, Erikstrup C, Valentiner-Branth P, Lundgren J, Østergaard L, Tolstrup M. SARS-CoV-2 vaccine-induced antibodies protect against Omicron breakthrough infection. iScience 2023; 26:107621. [PMID: 37682631 PMCID: PMC10481355 DOI: 10.1016/j.isci.2023.107621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/17/2023] [Accepted: 08/10/2023] [Indexed: 09/10/2023] Open
Abstract
SARS-CoV-2 Omicron quickly spread globally, also in regions with high vaccination coverage, emphasizing the importance of exploring the immunological requirements for protection against Omicron breakthrough infection. The test-negative matched case-control study (N = 964) characterized Omicron breakthrough infections in triple-vaccinated individuals from the ENFORCE cohort. Within 60 days before a PCR test spike-specific IgG levels were significantly lower in cases compared to controls (GMR [95% CI] for BA.2: 0.83 [0.73-0.95], p = 0.006). Multivariable logistic regression showed significant associations between high antibody levels and lower odds of infection (aOR [95% CI] for BA.2 spike-specific IgG: 0.65 [0.48-0.88], p = 0.006 and BA.2 ACE2-blocking antibodies: 0.46 [0.30-0.69], p = 0.0002). A sex-stratified analysis showed more pronounced associations for females than males. High levels of vaccine-induced antibodies provide partial protection against Omicron breakthrough infections. This is important knowledge to further characterize a threshold for protection against new variants and to estimate the necessity and timing of booster vaccination.
Collapse
Affiliation(s)
- Eva A.M. Baerends
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Astrid K. Hvidt
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Joanne Reekie
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ole S. Søgaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Nina B. Stærke
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Dorthe Raben
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Nielsen
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Kristine T. Petersen
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
| | - Maria R. Juhl
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
| | - Isik S. Johansen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Susan O. Lindvig
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Lone W. Madsen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Lothar Wiese
- Department of Medicine, Zealand University Hospital, Roskilde, Denmark
| | - Lene S. Knudsen
- Department of Medicine, Zealand University Hospital, Roskilde, Denmark
| | - Mette B. Iversen
- Department of Medicine, Zealand University Hospital, Roskilde, Denmark
| | - Thomas Benfield
- Department of Infectious Diseases, Copenhagen University Hospital – Amager and Hvidovre, Hvidovre, Denmark
- Departments of Clinical Medicine and Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Kasper K. Iversen
- Department of Infectious Diseases, Copenhagen University Hospital – Amager and Hvidovre, Hvidovre, Denmark
- Department of Cardiology and Emergency Medicine, Herlev Hospital, Herlev, Denmark
| | - Sidsel D. Andersen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anna K. Juhl
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lisa L. Dietz
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Signe R. Andreasen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Thea K. Fischer
- Departments of Clinical Medicine and Public Health, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Research, Nordsjællands University Hospital, Hillerød, Denmark
| | - Christian Erikstrup
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Palle Valentiner-Branth
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
| | - Jens Lundgren
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Departments of Clinical Medicine and Public Health, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
| | - Lars Østergaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
70
|
Pérez-Alós L, Hansen CB, Almagro Armenteros JJ, Madsen JR, Heftdal LD, Hasselbalch RB, Pries-Heje MM, Bayarri-Olmos R, Jarlhelt I, Hamm SR, Møller DL, Sørensen E, Ostrowski SR, Frikke-Schmidt R, Hilsted LM, Bundgaard H, Nielsen SD, Iversen KK, Garred P. Previous immunity shapes immune responses to SARS-CoV-2 booster vaccination and Omicron breakthrough infection risk. Nat Commun 2023; 14:5624. [PMID: 37699890 PMCID: PMC10497567 DOI: 10.1038/s41467-023-41342-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023] Open
Abstract
The heterogeneity of the SARS-CoV-2 immune responses has become considerably more complex over time and diverse immune imprinting is observed in vaccinated individuals. Despite vaccination, following the emergence of the Omicron variant, some individuals appear more susceptible to primary infections and reinfections than others, underscoring the need to elucidate how immune responses are influenced by previous infections and vaccination. IgG, IgA, neutralizing antibodies and T-cell immune responses in 1,325 individuals (955 of which were infection-naive) were investigated before and after three doses of the BNT162b2 vaccine, examining their relation to breakthrough infections and immune imprinting in the context of Omicron. Our study shows that both humoral and cellular responses following vaccination were generally higher after SARS-CoV-2 infection compared to infection-naive. Notably, viral exposure before vaccination was crucial to achieving a robust IgA response. Individuals with lower IgG, IgA, and neutralizing antibody responses postvaccination had a significantly higher risk of reinfection and future Omicron infections. This was not observed for T-cell responses. A primary infection before Omicron and subsequent reinfection with Omicron dampened the humoral and cellular responses compared to a primary Omicron infection, consistent with immune imprinting. These results underscore the significant impact of hybrid immunity for immune responses in general, particularly for IgA responses even after revaccination, and the importance of robust humoral responses in preventing future infections.
Collapse
Affiliation(s)
- Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Cecilie Bo Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Johannes Roth Madsen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Line Dam Heftdal
- Viro-immunology Research Unit, Department of Infectious Diseases, Section 8632, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Haematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Rasmus Bo Hasselbalch
- Department of Cardiology, Copenhagen University Hospital Herlev and Gentofte, Copenhagen, Denmark
- Department of Emergency Medicine, Copenhagen University Hospital Herlev and Gentofte, Copenhagen, Denmark
| | - Mia Marie Pries-Heje
- The Heart Center, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Rafael Bayarri-Olmos
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Recombinant Protein and Antibody Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ida Jarlhelt
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Sebastian Rask Hamm
- Viro-immunology Research Unit, Department of Infectious Diseases, Section 8632, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Dina Leth Møller
- Viro-immunology Research Unit, Department of Infectious Diseases, Section 8632, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Erik Sørensen
- Department of Clinical Immunology, Section 2034, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Section 2034, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Linda Maria Hilsted
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Henning Bundgaard
- The Heart Center, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Dam Nielsen
- Viro-immunology Research Unit, Department of Infectious Diseases, Section 8632, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kasper Karmark Iversen
- Department of Cardiology, Copenhagen University Hospital Herlev and Gentofte, Copenhagen, Denmark
- Department of Emergency Medicine, Copenhagen University Hospital Herlev and Gentofte, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
71
|
Rafie RA, Azimi L, Armin S, Aghamohammadi A, Karimi A, Fallah F, Khodaei H, Mansour Ghanaie R, Alebouyeh M. Evaluation of Covid-19 anti-spike IgG antibody five months after the second Covid-19 vaccination. GMS HYGIENE AND INFECTION CONTROL 2023; 18:Doc20. [PMID: 37829253 PMCID: PMC10566013 DOI: 10.3205/dgkh000446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Background Studies in different communities have shown significant differences in IgG antibody titers in the time period after the first and second doses of the vaccines. This study aimed to serologically evaluate the IgG anti-spike antibody titer five months after injection of the second COVID-19 vaccine in healthcare workers. Materials and method This study was performed in healthcare personnel for whom five months had passed since their second anti-Covid-19 vaccination. The level of IgG antibody against SARS-CoV-2 spike protein was measured by ELISA. Healthcare workers in Mofid Children's hospital received three brands of vaccines: Sputnik V, Sinopharm, and AstraZeneca. Results The mean titer of anti-spike IgG was 4.3±2.29 units. The percentage of positive cases of the antibody was estimated to be 96.4%. The titer of anti-spike IgG antibody was dependent on both the occupational area and a positive history of Covid-19 disease. Conclusion About 96.4% of the staff vaccinated against Covid-19 had a high titer of anti-spike IgG antibody even five months after inoculation of the second dose. The field of occupational can affect the level of antibody present.
Collapse
Affiliation(s)
- Reyhaneh Alipoor Rafie
- Department of Infectious Diseases, Shahid Beheshti University of Medical sciences, Tehran, Iran
- Pediatric Infections Research Center, Research Institute for Children’s Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Azimi
- Pediatric Infections Research Center, Research Institute for Children’s Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahnaz Armin
- Pediatric Infections Research Center, Research Institute for Children’s Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirali Aghamohammadi
- Pediatric Infections Research Center, Research Institute for Children’s Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdollah Karimi
- Pediatric Infections Research Center, Research Institute for Children’s Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Fallah
- Pediatric Infections Research Center, Research Institute for Children’s Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hannan Khodaei
- Pediatric Infections Research Center, Research Institute for Children’s Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roxana Mansour Ghanaie
- Pediatric Infections Research Center, Research Institute for Children’s Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Alebouyeh
- Pediatric Infections Research Center, Research Institute for Children’s Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
72
|
Hergenroeder GE, Faino AV, Cogen JD, Genatossio A, McNamara S, Pascual M, Hernandez RE. Seroprevalence and clinical characteristics of SARS-CoV-2 infection in children with cystic fibrosis. Pediatr Pulmonol 2023; 58:2478-2486. [PMID: 37314149 PMCID: PMC11548890 DOI: 10.1002/ppul.26528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/21/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND People with cystic fibrosis (PwCF) have chronic lung disease and may be at increased risk of coronavirus disease 2019 (COVID-19)-related morbidity and mortality. This study aimed to determine seroprevalence and clinical characteristics of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in children with cystic fibrosis (CF), and to assess antibody responses following SARS-CoV-2 infection or vaccination. METHODS Children and adolescents with CF followed at Seattle Children's Hospital were enrolled between July 20, 2020 and February 28, 2021. SARS-CoV-2 serostatus was determined on enrollment at 6 and 11 months (±2 months) for nucleocapsid and spike IgG. Participants completed intake and weekly surveys inquiring about SARS-CoV-2 exposures, viral/respiratory illnesses, and symptoms. RESULTS Of 125 PwCF enrolled, 14 (11%) had positive SARS-CoV-2 antibodies consistent with recent or past infection. Seropositive participants were more likely to identify as Hispanic (29% vs. 8%, p = 0.04) and have pulmonary exacerbations requiring oral antibiotics in the year prior (71% vs. 41%, p = 0.04). Five seropositive individuals (35.7%) were asymptomatic, while six (42.9%) reported mild symptoms, primarily cough and nasal congestion. Antispike protein IgG levels were approximately 10-fold higher in participants following vaccination compared with participants who had natural infection alone (p < 0.0001) and resembled levels previously reported in the general population. CONCLUSIONS A majority of PwCF have mild or no symptoms of SARS-CoV-2 making it difficult to distinguish from baseline respiratory symptoms. Hispanic PwCF may be disproportionately impacted, consistent with racial and ethnic COVID-19 disparities among the general US population. Vaccination in PwCF generated antibody responses similar to those previously reported in the general population.
Collapse
Affiliation(s)
- Georgene E. Hergenroeder
- Division of Pulmonary and Sleep Medicine, Seattle Children’s Hospital, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Anna V. Faino
- Core for Biostatistics, Epidemiology, and Analytics in Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Jonathan D. Cogen
- Division of Pulmonary and Sleep Medicine, Seattle Children’s Hospital, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Center for Clinical and Translational Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Alan Genatossio
- Division of Pulmonary and Sleep Medicine, Seattle Children’s Hospital, Seattle, Washington, USA
| | - Sharon McNamara
- Division of Pulmonary and Sleep Medicine, Seattle Children’s Hospital, Seattle, Washington, USA
| | - Michael Pascual
- Division of Pulmonary and Sleep Medicine, Seattle Children’s Hospital, Seattle, Washington, USA
| | - Rafael E. Hernandez
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Division of Infectious Diseases, Seattle Children’s Hospital, Seattle, Washington, USA
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| |
Collapse
|
73
|
Oliosi E, Flahault A, Charre C, Veyer D, Combier A, Lafont E, Karras A, Mouthon L, Avouac J, Terrier B, Hadjadj J. Impact of rituximab on humoral response to SARS-CoV-2 vaccination in previously vaccinated patients with autoimmune diseases. Clin Rheumatol 2023; 42:2485-2490. [PMID: 37243801 PMCID: PMC10224652 DOI: 10.1007/s10067-023-06638-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/16/2023] [Accepted: 05/12/2023] [Indexed: 05/29/2023]
Abstract
SARS-CoV-2 infection is more severe in patients undergoing rituximab (RTX) treatment. Humoral response to vaccination is severely impaired in patients already treated with RTX, but data on antibody persistence in patients initiating RTX are lacking. We evaluated the impact of RTX initiation on humoral response to SARS-CoV-2 vaccination in previously vaccinated patients with immune-mediated inflammatory diseases. We performed a retrospective, multicenter study evaluating the evolution of anti-spike antibodies and breakthrough infections after initiation of RTX in previously vaccinated patients with protective levels of anti-SARS-CoV-2 antibodies. Threshold for anti-S antibodies positivity and protection were 30 and 264 BAU/mL, respectively. We included 31 previously vaccinated patients initiating RTX (21 female, median age 57 years). At first RTX infusion, 12 (39%) patients had received 2 doses of vaccine, 15 (48%) had received 3 doses, and 4 (13%) had received 4 doses. The most frequent underlying diseases were ANCA-associated vasculitis (29%) and rheumatoid arthritis (23%). Median anti-S antibody titers at RTX initiation, 3 months, and 6 months were 1620 (589-2080), 1055 (467-2080), and 407 (186-659) BAU/mL, respectively. Overall, antibody titers waned by almost two-fold at 3 months and four-fold at 6 months. Median antibody titers were significantly higher in patients who received ≥3 doses compared to those who received only 2 doses. Three patients developed SARS-CoV-2 infection without any severe symptom. Anti-SARS-CoV-2 antibody titers in previously vaccinated patients decline after RTX initiation similarly to general population. Specific monitoring is useful to anticipate prophylactic strategies. Key Points • Anti-SARS-CoV-2 antibody titers in previously vaccinated patients decline after rituximab initiation similarly to the general population. • The number of dose of vaccine before rituximab initiation is associated with higher antibody titers at month 3. • Monitoring antibody levels is mandatory to initiate prophylactic strategies in this population.
Collapse
Affiliation(s)
- E Oliosi
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, Hôpital Cochin, Université de Paris Cité, 75014, Paris, France.
- Service de Maladies infectieuses et tropicales, Hôpital Bicêtre, APHP, 78 rue du Général Leclerc, 94270, Le Kremlin-Bicêtre, France.
| | - A Flahault
- Department of Nephrology, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - C Charre
- Department of Virology, AP-HP, Hôpital Cochin, 75014, Paris, France
| | - D Veyer
- Department of Virology, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - A Combier
- Department of Rheumatology, AP-HP, Hôpital Cochin, 75014, Paris, France
| | - E Lafont
- Department of Internal Medicine, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - A Karras
- Department of Nephrology, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - L Mouthon
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, Hôpital Cochin, Université de Paris Cité, 75014, Paris, France
| | - J Avouac
- Department of Rheumatology, AP-HP, Hôpital Cochin, 75014, Paris, France
| | - B Terrier
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, Hôpital Cochin, Université de Paris Cité, 75014, Paris, France
| | - J Hadjadj
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, Hôpital Cochin, Université de Paris Cité, 75014, Paris, France
| |
Collapse
|
74
|
Liu H, Aviszus K, Zelarney P, Liao SY, Gerber AN, Make B, Wechsler ME, Marrack P, Reinhardt RL. Vaccine-elicited B- and T-cell immunity to SARS-CoV-2 is impaired in chronic lung disease patients. ERJ Open Res 2023; 9:00400-2023. [PMID: 37583809 PMCID: PMC10423317 DOI: 10.1183/23120541.00400-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/24/2023] [Indexed: 08/17/2023] Open
Abstract
Background While vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) provides significant protection from coronavirus disease 2019, the protection afforded to individuals with chronic lung disease is less well established. This study seeks to understand how chronic lung disease impacts SARS-CoV-2 vaccine-elicited immunity. Methods Deep immune phenotyping of humoral and cell-mediated responses to the SARS-CoV-2 vaccine was performed in patients with asthma, COPD and interstitial lung disease (ILD) compared to healthy controls. Results 48% of vaccinated patients with chronic lung diseases had reduced antibody titres to the SARS-CoV-2 vaccine antigen relative to healthy controls. Vaccine antibody titres were significantly reduced among asthma (p<0.035), COPD (p<0.022) and a subset of ILD patients as early as 3-4 months after vaccination, correlating with decreased vaccine-specific memory B-cells in circulation. Vaccine-specific memory T-cells were significantly reduced in patients with asthma (CD8+ p<0.004; CD4+ p<0.023) and COPD (CD8+ p<0.008) compared to healthy controls. Impaired T-cell responsiveness was also observed in a subset of ILD patients (CD8+ 21.4%; CD4+ 42.9%). Additional heterogeneity between healthy and disease cohorts was observed among bulk and vaccine-specific follicular T-helper cells. Conclusions Deep immune phenotyping of the SARS-CoV-2 vaccine response revealed the complex nature of vaccine-elicited immunity and highlights the need for more personalised vaccination schemes in patients with underlying lung conditions.
Collapse
Affiliation(s)
- Haolin Liu
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
| | - Katja Aviszus
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
| | | | - Shu-Yi Liao
- Department of Medicine, National Jewish Health, Denver, CO, USA
- Division of Environmental and Occupational Health Sciences, National Jewish Health, Denver, CO, USA
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Anthony N. Gerber
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
- Department of Medicine, National Jewish Health, Denver, CO, USA
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - Barry Make
- Department of Medicine, National Jewish Health, Denver, CO, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - Michael E. Wechsler
- Department of Medicine, National Jewish Health, Denver, CO, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - Philippa Marrack
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - R. Lee Reinhardt
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
75
|
Song W, Fang Z, Ma F, Li J, Huang Z, Zhang Y, Li J, Chen K. The role of SARS-CoV-2 N protein in diagnosis and vaccination in the context of emerging variants: present status and prospects. Front Microbiol 2023; 14:1217567. [PMID: 37675423 PMCID: PMC10478715 DOI: 10.3389/fmicb.2023.1217567] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/31/2023] [Indexed: 09/08/2023] Open
Abstract
Despite many countries rapidly revising their strategies to prevent contagions, the number of people infected with Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to surge. The emergent variants that can evade the immune response significantly affect the effectiveness of mainstream vaccines and diagnostic products based on the original spike protein. Therefore, it is essential to focus on the highly conserved nature of the nucleocapsid protein as a potential target in the field of vaccines and diagnostics. In this regard, our review initially discusses the structure, function, and mechanism of action of N protein. Based on this discussion, we summarize the relevant research on the in-depth development and application of diagnostic methods and vaccines based on N protein, such as serology and nucleic acid detection. Such valuable information can aid in designing more efficient diagnostic and vaccine tools that could help end the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Wanchen Song
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhongbiao Fang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Feike Ma
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jiaxuan Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Zhiwei Huang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yanjun Zhang
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Jianhua Li
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| |
Collapse
|
76
|
Roy A, Saade C, Josset L, Clément B, Morfin F, Destras G, Valette M, Icard V, Billaud G, Oblette A, Debombourg M, Garrigou C, Brengel-Pesce K, Generenaz L, Saker K, Hernu R, Pozzetto B, Lina B, Trabaud MA, Trouillet-Assant S, Bal A. Determinants of protection against SARS-CoV-2 Omicron BA.1 and Delta infections in fully vaccinated outpatients. J Med Virol 2023; 95:e28984. [PMID: 37503561 DOI: 10.1002/jmv.28984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/23/2023] [Accepted: 07/13/2023] [Indexed: 07/29/2023]
Abstract
We aimed to evaluate the association between the humoral and cellular immune responses and symptomatic SARS-CoV-2 infection with Delta or Omicron BA.1 variants in fully vaccinated outpatients. Anti-receptor binding domain (RBD) IgG levels and interferon-gamma (IFN-γ) release were evaluated at PCR-diagnosis of SARS-CoV-2 in 636 samples from negative and positive patients during Delta and Omicron BA.1 periods. Median levels of anti-RBD IgG in positive patients were significantly lower than in negative patients for both variants (p < 0.05). The frequency of Omicron BA.1 infection in patients with anti-RBD IgG concentrations ≥1000 binding antibody units (BAU)/mL was 51.0% and decreased to 34.4% in patients with concentrations ≥3000 BAU/mL. For Delta infection, the frequency of infection was significantly lower when applying the same anti-RBD IgG thresholds (13.3% and 5.3% respectively, p < 0.05). In addition, individuals in the hybrid immunity group had a 4.5 times lower risk of Delta infection compared to the homologous vaccination group (aOR = 0.22, 95% CI: [0.05-0.64]. No significant decrease in the risk of Omicron BA.1 infection was observed in the hybrid group compared to the homologous group, but the risk decreased within the hybrid group as anti-RBD IgG titers increased (aOR = 0.08, 95% CI: [0.01-0.41], p = 0.008). IFN-γ release post-SARS-CoV-2 peptide stimulation was not different between samples from patients infected (either with Delta or Omicron BA.1 variant) or not (p > 0.05). Our results show that high circulating levels of anti-RBD IgG and hybrid immunity were independently associated with a lower risk of symptomatic SARS-CoV-2 infection in outpatients with differences according to the infecting variant (www.clinicaltrials.gov; ID NCT05060939).
Collapse
Affiliation(s)
- Alvaro Roy
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
- ECDC Fellowship Programme, Public Health Microbiology Path (EUPHEM), European Centre for Disease Prevention and Control (ECDC), Solna, Sweden
| | - Carla Saade
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- Joint Research Unit Civils Hospices of Lyon-bioMérieux, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Laurence Josset
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- GenEPII Sequencing Platform, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Bénédicte Clément
- Services des urgences, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Florence Morfin
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- GenEPII Sequencing Platform, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Grégory Destras
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- GenEPII Sequencing Platform, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Martine Valette
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
| | - Vinca Icard
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
| | - Geneviéve Billaud
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
| | - Antoine Oblette
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
- GenEPII Sequencing Platform, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Marion Debombourg
- Joint Research Unit Civils Hospices of Lyon-bioMérieux, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Christine Garrigou
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
| | - Karen Brengel-Pesce
- Joint Research Unit Civils Hospices of Lyon-bioMérieux, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Laurence Generenaz
- Joint Research Unit Civils Hospices of Lyon-bioMérieux, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Kahina Saker
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
- Joint Research Unit Civils Hospices of Lyon-bioMérieux, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Romain Hernu
- Services des urgences, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Bruno Pozzetto
- Team GIMAP, CIRI-Centre International de Recherche en Infectiologie, Université Jean Monnet de Saint-Etienne, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, Saint-Etienne, France
- Laboratoire des Agents Infectieux et d'Hygiène, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Etienne, France
| | - Bruno Lina
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- GenEPII Sequencing Platform, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Mary-Anne Trabaud
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
| | - Sophie Trouillet-Assant
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- Joint Research Unit Civils Hospices of Lyon-bioMérieux, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Antonin Bal
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des virus des infections respiratoires, Hospices Civils de Lyon, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- GenEPII Sequencing Platform, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
77
|
Seekircher L, Bánki Z, Kimpel J, Rössler A, Schäfer H, Falkensammer B, Bante D, Forer L, Schönherr S, Harthaller T, Sacher M, Ower C, Tschiderer L, Ulmer H, Krammer F, von Laer D, Borena W, Willeit P. Immune response after two doses of the BNT162b2 COVID-19 vaccine and risk of SARS-CoV-2 breakthrough infection in Tyrol, Austria: an open-label, observational phase 4 trial. THE LANCET. MICROBE 2023; 4:e612-e621. [PMID: 37354911 PMCID: PMC10284585 DOI: 10.1016/s2666-5247(23)00107-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 02/03/2023] [Accepted: 03/06/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Correlates of protection could help to assess the extent to which a person is protected from SARS-CoV-2 infection after vaccination (so-called breakthrough infection). We aimed to clarify associations of antibody and T-cell responses after vaccination against COVID-19 with risk of a SARS-CoV-2 breakthrough infection and whether measurement of these responses enhances risk prediction. METHODS We did an open-label, phase 4 trial in two community centres in the Schwaz district of the Federal State of Tyrol, Austria, before the emergence of the omicron (B.1.1.529) variant of SARS-CoV-2. We included individuals (aged ≥16 years) a mean of 35 days (range 27-43) after they had received a second dose of the BNT162b2 (Pfizer-BioNTech) COVID-19 vaccine. We quantified associations between immunological parameters and breakthrough infection and assessed whether information on these parameters improves risk discrimination. The study is registered with the European Union Drug Regulating Authorities Clinical Trials Database, 2021-002030-16. FINDINGS 2760 individuals (1682 [60·9%] female, 1078 [39·1%] male, mean age 47·4 years [SD 14·5]) were enrolled into this study between May 15 and May 21, 2021, 712 (25·8%) of whom had a previous SARS-CoV-2 infection. Over a median follow-up of 5·9 months, 68 (2·5%) participants had a breakthrough infection. In models adjusted for age, sex, and previous infection, hazard ratios for breakthrough infection for having twice the immunological parameter level at baseline were 0·72 (95% CI 0·60-0·86) for anti-spike IgG, 0·80 (0·70-0·92) for neutralising antibodies in a surrogate virus neutralisation assay, 0·84 (0·58-1·21) for T-cell response after stimulation with a CD4 peptide pool, and 0·77 (0·54-1·08) for T-cell response after stimulation with a combined CD4 and CD8 peptide pool. For neutralising antibodies measured in a nested case-control sample using a pseudotyped virus neutralisation assay, the corresponding odds ratio was 0·78 (0·62-1·00). Among participants with previous infection, the corresponding hazard ratio was 0·73 (0·61-0·88) for anti-nucleocapsid Ig. Addition of anti-spike IgG information to a model containing information on age and sex improved the C-index by 0·085 (0·027-0·143). INTERPRETATION In contrast to T-cell response, higher levels of binding and neutralising antibodies were associated with a reduced risk of breakthrough SARS-CoV-2 infection. The assessment of anti-spike IgG enhances the prediction of incident breakthrough SARS-CoV-2 infection and could therefore be a suitable correlate of protection in practice. Our phase 4 trial measured both humoral and cellular immunity and had a 6-month follow-up period; however, the longer-term protection against emerging variants of SARS-CoV-2 remains unclear. FUNDING None.
Collapse
Affiliation(s)
- Lisa Seekircher
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria
| | - Zoltán Bánki
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Janine Kimpel
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Annika Rössler
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Helena Schäfer
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - David Bante
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Forer
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Teresa Harthaller
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Magdalena Sacher
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Cornelia Ower
- Department of Surgery, University Hospital of Trauma Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Lena Tschiderer
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria
| | - Hanno Ulmer
- Institute of Medical Statistics and Informatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Krammer
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dorothee von Laer
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Wegene Borena
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria.
| | - Peter Willeit
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria; Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Vienna, Austria; Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| |
Collapse
|
78
|
De Pace V, Bruzzone B, Ricucci V, Calcavecchia N, Guarona G, Giberti I, Costa E, Ogliastro M, Galano B, Nigro N, Murgia D, Nanni L, Orsi A. Long follow-up of BNT162b2 mRNA vaccine in healthcare workers (2020-2022): A retrospective longitudinal SARS-CoV-2 serological surveillance. Hum Vaccin Immunother 2023; 19:2258632. [PMID: 37724517 PMCID: PMC10512804 DOI: 10.1080/21645515.2023.2258632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/11/2023] [Indexed: 09/21/2023] Open
Abstract
SARS-CoV-2 anti-spike IgG production and protection from severe respiratory illness should be explored in greater depth after COVID-19 booster vaccination. This longitudinal observational retrospective study investigated the anti-spike IgG response elicited by the first, second and booster doses of BNT162b2 mRNA vaccine in healthcare workers (HCW) at San Martino IRCCS Policlinico Hospital (Genoa) up to the 12th month. Sequential blood sampling was performed at T0 (prior to vaccination), T1 (21 days after the 1st dose of vaccine), T2, T3, T4, T5, T6 (7 days and 1, 3, 6 and 9 months after the 2nd dose, respectively), T7 and T8 (1 and 3 months after a booster dose). A SARS-CoV-2 IgG panel (Bio-Rad, Marnes-la-Coquette, France) was used to determine levels of receptor-binding domain (RBD), spike-1 (S1), spike-2 and nucleocapsid structural proteins of SARS-CoV-2. In the 51 HCWs evaluated, seroprevalence was 96% (49/51) at T1 and 100% (51/51) from T2 to T5 for RBD and S1. At T6, only one HCW was negative. T2 [RBD = 2945 (IQR:1693-5364); S1 = 1574 (IQR:833-3256) U/mL], and T7 [RBD = 8204 (IQR:4129-11,912); S1 = 4124 (IQR:2124-6326) U/mL] were characterized by the highest antibody values. Significant humoral increases in RBD and S1 were documented at T7 and T8 compared to T2 and T4, respectively (p-value < .001). Following vaccination with BNT162b2 and a booster dose in the 9th month, naïve and healthy subjects show high antibody titers up to 12 months and a protective humoral response against COVID-19 disease lasting up to 20 months after the last booster.
Collapse
Affiliation(s)
- Vanessa De Pace
- Hygiene Unit, San Martino Policlinico Hospital - IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Bianca Bruzzone
- Hygiene Unit, San Martino Policlinico Hospital - IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Valentina Ricucci
- Hygiene Unit, San Martino Policlinico Hospital - IRCCS for Oncology and Neurosciences, Genoa, Italy
| | | | - Giulia Guarona
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Irene Giberti
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Elisabetta Costa
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | | | - Barbara Galano
- Hygiene Unit, San Martino Policlinico Hospital - IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Nicola Nigro
- Hygiene Unit, San Martino Policlinico Hospital - IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Daniele Murgia
- Medicine Laboratory, San Martino Policlinico Hospital - IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Luca Nanni
- Medicine Laboratory, San Martino Policlinico Hospital - IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Andrea Orsi
- Hygiene Unit, San Martino Policlinico Hospital - IRCCS for Oncology and Neurosciences, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| |
Collapse
|
79
|
Hertz T, Levy S, Ostrovsky D, Oppenheimer H, Zismanov S, Kuzmina A, Friedman LM, Trifkovic S, Brice D, Chun-Yang L, Cohen-Lavi L, Shemer-Avni Y, Cohen-Lahav M, Amichay D, Keren-Naus A, Voloshin O, Weber G, Najjar-Debbiny R, Chazan B, McGargill MA, Webby R, Chowers M, Novack L, Novack V, Taube R, Nesher L, Weinstein O. Correlates of protection for booster doses of the SARS-CoV-2 vaccine BNT162b2. Nat Commun 2023; 14:4575. [PMID: 37516771 PMCID: PMC10387073 DOI: 10.1038/s41467-023-39816-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 06/28/2023] [Indexed: 07/31/2023] Open
Abstract
Vaccination, especially with multiple doses, provides substantial population-level protection against COVID-19, but emerging variants of concern (VOC) and waning immunity represent significant risks at the individual level. Here we identify correlates of protection (COP) in a multicenter prospective study following 607 healthy individuals who received three doses of the Pfizer-BNT162b2 vaccine approximately six months prior to enrollment. We compared 242 individuals who received a fourth dose to 365 who did not. Within 90 days of enrollment, 239 individuals contracted COVID-19, 45% of the 3-dose group and 30% of the four-dose group. The fourth dose elicited a significant rise in antibody binding and neutralizing titers against multiple VOCs reducing the risk of symptomatic infection by 37% [95%CI, 15%-54%]. However, a group of individuals, characterized by low baseline titers of binding antibodies, remained susceptible to infection despite significantly increased neutralizing antibody titers upon boosting. A combination of reduced IgG levels to RBD mutants and reduced VOC-recognizing IgA antibodies represented the strongest COP in both the 3-dose group (HR = 6.34, p = 0.008) and four-dose group (HR = 8.14, p = 0.018). We validated our findings in an independent second cohort. In summary combination IgA and IgG baseline binding antibody levels may identify individuals most at risk from future infections.
Collapse
Affiliation(s)
- Tomer Hertz
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
- National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Research Center, Seattle, USA.
| | - Shlomia Levy
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Daniel Ostrovsky
- Clinical Research Center, Soroka University Medical Center, and the faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hanna Oppenheimer
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Shosh Zismanov
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alona Kuzmina
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Lilach M Friedman
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sanja Trifkovic
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - David Brice
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Lin Chun-Yang
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Liel Cohen-Lavi
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yonat Shemer-Avni
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Laboratory of Virology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Merav Cohen-Lahav
- Laboratory of Management, Soroka University Medical Center, Beer-Sheva, Israel
| | - Doron Amichay
- Central Laboratory, Clalit Health Services & Dept. of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheba, Israel
| | - Ayelet Keren-Naus
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Laboratory of Virology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Olga Voloshin
- Laboratory of Virology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Gabriel Weber
- Infectious Diseases Unit, Lady Davis Carmel Medical Center, Haifa, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ronza Najjar-Debbiny
- Infectious Diseases Unit, Lady Davis Carmel Medical Center, Haifa, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Bibiana Chazan
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Infectious Diseases Unit, Emek Medical Center, Afula, Israel
| | - Maureen A McGargill
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Richard Webby
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Michal Chowers
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Meir Medical Center, Kfar Saba, Israel
| | - Lena Novack
- Clinical Research Center, Soroka University Medical Center, and the faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Victor Novack
- Clinical Research Center, Soroka University Medical Center, and the faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ran Taube
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Lior Nesher
- Infectious Disease Institute, Soroka University Medical Center, and Faculty of Health Sciences, Ben-Gurion University, Beer Sheba, Israel.
| | - Orly Weinstein
- Dept. of Health systems management, faculty of health sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Hospital division, Clalit Health Services, Tel Aviv, Israel
| |
Collapse
|
80
|
Barda N, Lustig Y, Indenbaum V, Zibly D, Joseph G, Asraf K, Weiss-Ottolenghi Y, Amit S, Kliker L, Abd Elkader B, Ben-Ami E, Canetti M, Koren R, Katz-Likvornik S, Halpern O, Mendelson E, Doolman R, Harats D, Kreiss Y, Mandelboim M, Regev-Yochay G. Immunogenicity of Omicron BA.1-adapted BNT162b2 vaccines: randomized trial, 3-month follow-up. Clin Microbiol Infect 2023; 29:918-923. [PMID: 36921715 PMCID: PMC10010049 DOI: 10.1016/j.cmi.2023.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/02/2023] [Accepted: 03/05/2023] [Indexed: 03/16/2023]
Abstract
OBJECTIVES The capability of the SARS-CoV-2 Omicron variant to escape immunity conferred by mRNA vaccines has led to the development of Omicron-adapted vaccines. In this study, we aimed to compare the immune response with the ancestral strain and with the BA.1 Omicron variant after administration of the original vaccine and the Omicron-adapted vaccine. METHODS This is an ongoing phase 3, double-blinded randomized controlled trial, comparing the original BNT161b2 vaccine, monovalent Omicron BA.1-adapted BNT161b2 vaccine, and bivalent combinations. Each vaccine was given at a 30 μg and 60 μg dose. Primary outcomes considered included neutralization titers of SARS-CoV-2 ancestral strain and Omicron BA.1. Exploratory endpoints included neutralization titers for Omicron BA.5, and the incidence of COVID-19 cases. RESULTS Overall, 122 individuals (22, 19, 20, 20, 20, 20, and 21 in each arm) completed a 90-day follow-up. Three months after vaccination, adjusting for baseline levels, neutralizing antibody titers were 0.63 (95% CI: 0.3-1.32) and 0.54 (0.24-1.2) for monovalent/60 μg, 0.9 (0.42-1.92) and 2.69 (1.17-6.17) times for monovalent-Omi.BA.1/30 μg, 1.28 (0.6-2.75) and 2.79 (1.21-6.41) times for monovalent-Omi.BA.1/60 μg, 0.96 (0.46-1.97) and 2.07 (0.93-4.58) times for bivalent-Omi.BA.1/30 μg, and 0.79 (0.38-1.63) and 1.95 (0.88-4.32) times for bivalent-Omi.BA.1/60 μg when compared with BNT162b2/30 μg against the ancestral strain and BA.1 variant, respectively. DISCUSSION BA.1-adapted mRNA vaccines lead to a stronger neutralizing antibody response against the Omicron BA.1 sub-variant.
Collapse
Affiliation(s)
- Noam Barda
- ARC Innovation Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Software and Information Systems Engineering, Ben-Gurion University of the Negev, Be'er Sheva, Israel; Epidemiology, Biostatistics and Community Health Services, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Yaniv Lustig
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Central Virology Laboratory, Public Health Services, Ministry of Health, Tel Hashomer, Ramat Gan, Israel
| | - Victoria Indenbaum
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Central Virology Laboratory, Public Health Services, Ministry of Health, Tel Hashomer, Ramat Gan, Israel
| | - Daniel Zibly
- The Infection Prevention & Control Unit, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Gili Joseph
- The Infection Prevention & Control Unit, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Keren Asraf
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; The Dworman Automated-Mega Laboratory, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Yael Weiss-Ottolenghi
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; The Infection Prevention & Control Unit, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Sharon Amit
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Clinical Microbiology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Limor Kliker
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Central Virology Laboratory, Public Health Services, Ministry of Health, Tel Hashomer, Ramat Gan, Israel
| | - Bayan Abd Elkader
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Central Virology Laboratory, Public Health Services, Ministry of Health, Tel Hashomer, Ramat Gan, Israel
| | - Eytan Ben-Ami
- Phase 1 Clinical Trials Unit, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Michal Canetti
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; The Infection Prevention & Control Unit, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Ravit Koren
- Central Virology Laboratory, Public Health Services, Ministry of Health, Tel Hashomer, Ramat Gan, Israel
| | - Shiri Katz-Likvornik
- Central Virology Laboratory, Public Health Services, Ministry of Health, Tel Hashomer, Ramat Gan, Israel
| | - Osnat Halpern
- Central Virology Laboratory, Public Health Services, Ministry of Health, Tel Hashomer, Ramat Gan, Israel
| | - Ella Mendelson
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Central Virology Laboratory, Public Health Services, Ministry of Health, Tel Hashomer, Ramat Gan, Israel
| | - Ram Doolman
- The Dworman Automated-Mega Laboratory, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Dror Harats
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; General Management, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Yitshak Kreiss
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; General Management, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Michal Mandelboim
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Central Virology Laboratory, Public Health Services, Ministry of Health, Tel Hashomer, Ramat Gan, Israel
| | - Gili Regev-Yochay
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; The Infection Prevention & Control Unit, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.
| |
Collapse
|
81
|
Razu MH, Ahmed ZB, Hossain MI, Rabby MRI, Akter F, Karmaker P, Khan MR, Moniruzzaman M, Khan M. SARS CoV-2 IgG positivity among the people in Dhaka city: An observation from the post vaccine period. Heliyon 2023; 9:e17767. [PMID: 37501992 PMCID: PMC10303751 DOI: 10.1016/j.heliyon.2023.e17767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023] Open
Abstract
Immunity status after mass vaccination program against SARS CoV-2 has not been evaluated in Bangladesh. This study aims to assess the IgG response against SARS-CoV-2 among the vaccine receivers in Bangladesh. After signed consent, blood samples were tested for SARS CoV-2 IgG from volunteers between March, 21 and April, 22 using ELISA where IgG index ≥0.9 was considered as positive Among 3034 participants, IgG positivity was calculated approximately 82% for vaccine recipients; lowest (58%) during March-April, 21 which increased to 85-95% later. IgG positivity and mean index was 82% and 3.04 in vaccinated whereas 56% and 1.5 in unvaccinated cases. IgG positivity and mean index reduced with age: 90% and 2.56, 79% and 2.23, 73% and 2.13 in 18-40 y, 41-60 y, >60 y group respectively. Vaccinated with COVID-19 history showed highest IgG positivity and index (94% and 3.1) compared to vaccinated without COVID-19 history (76% and 1.6), unvaccinated with COVID-19 history (75% and 1.5) and unvaccinated without COVID-19 history (51% and 0.9). IgG positivity and index reduced as interval between IgG testing and vaccination increases. Our findings suggest a robust IgG response among the vaccine recipients. Negative correlation of IgG positivity and index with age and time necessitates continuous monitoring of immunity status.
Collapse
|
82
|
Golec M, Zembala-John J, Fronczek M, Konka A, Bochenek A, Wystyrk K, Botor H, Zalewska M, Chrapiec M, Kasperczyk S, Brzoza Z, Bułdak RJ. Relationship between anthropometric and body composition parameters and anti-SARS-CoV-2 specific IgG titers in females vaccinated against COVID-19 according to the heterologous vaccination course: A cohort study. PLoS One 2023; 18:e0287128. [PMID: 37310975 DOI: 10.1371/journal.pone.0287128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
INTRODUCTION The aim of this cohort study was to evaluate the relationship between anthropometric and body composition parameters and anti-SARS-CoV-2 IgG titers in a group of females who were vaccinated against COVID-19 with two doses of ChAdOx1 vaccine and then boosted with the BNT162b2 vaccine. MATERIALS AND METHODS The study group consisted of 63 women. Basic demographic and clinical data were collected. To assess the anti-SARS-CoV-2 immunoglobulin G titers following the vaccination, five blood draws were performed: 1) before the first dose, 2) before the second dose, 3) 14-21 days after the primary vaccination, 4) before the booster, and 5) 21 days after the booster. Blood samples were analyzed using a two-step enzymatic chemiluminescent assay. Body mass index and body composition were evaluated using bioelectrical impedance analysis. To select the most distinguishing parameters and correlations between anthropometric and body composition parameters and anti-SARS-CoV-2 IgG titers, factor analysis using the Principal Component Analysis was conducted. RESULTS Sixty-three females (mean age: 46.52 years) who met the inclusion criteria were enrolled. 40 of them (63.50%) participated in the post-booster follow-up. After receiving two doses of the ChAdOx1 vaccine, the study group's anti-SARS-CoV-2 IgG titers were 67.19 ± 77.44 AU/mL (mean ± SD), whereas after receiving a heterologous mRNA booster, the level of anti-SARS-CoV-2 IgG titers was about three-times higher and amounted to 212.64 ± 146.40 AU/mL (mean ± SD). Our data shows that seropositivity, obesity, non-fat-related, and fat-related body composition parameters all had a significant effect on the level of IgG titer after a two-dose vaccination of ChAdOx1. However, only non-fat-related and fat-related body composition parameters had a significant effect on the IgG titer after booster vaccination. CONCLUSION COVID-19 infection before the first dose of vaccination is not related to IgG titer after booster administration. Body composition has a significant effect on the production of anti-SARS-CoV-2 IgG after booster vaccination in females.
Collapse
Affiliation(s)
- Marlena Golec
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
| | - Joanna Zembala-John
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
- Department of Medicine and Environmental Epidemiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland
- Silesian Center for Heart Diseases, Zabrze, Poland
| | - Martyna Fronczek
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Adam Konka
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
| | - Aneta Bochenek
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
| | - Karolina Wystyrk
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
| | | | - Marzena Zalewska
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
- Department of Basic Medical Sciences, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Bytom, Poland
| | - Martyna Chrapiec
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
| | - Sławomir Kasperczyk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Zenon Brzoza
- Department of Internal Diseases, Allergology, Endocrinology and Gastroenterology, Institute of Medical Sciences, University of Opole, Opole, Poland
| | - Rafał J Bułdak
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Opole, Poland
| |
Collapse
|
83
|
DeJonge PM, Lambrou AS, Segaloff HE, Bateman A, Sterkel A, Griggs C, Baggott J, Kelly P, Thornburg N, Epperson M, Desamu-Thorpe R, Abedi G, Hsu CH, Nakayama JY, Ruffin J, Turner-Harper D, Matanock A, Almendares O, Whaley M, Chakrabarti A, DeGruy K, Daly M, Westergaard R, Tate JE, Kirking HL. Assessment of Anti-SARS-CoV-2 antibody levels among university students vaccinated with different COVID-19 primary and booster doses - fall 2021, Wisconsin. BMC Infect Dis 2023; 23:374. [PMID: 37277736 DOI: 10.1186/s12879-023-08332-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/16/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND University students commonly received COVID-19 vaccinations before returning to U.S. campuses in the Fall of 2021. Given likely immunologic variation among students based on differences in type of primary series and/or booster dose vaccine received, we conducted serologic investigations in September and December 2021 on a large university campus in Wisconsin to assess anti-SARS-CoV-2 antibody levels. METHODS We collected blood samples, demographic information, and COVID-19 illness and vaccination history from a convenience sample of students. Sera were analyzed for both anti-spike (anti-S) and anti-nucleocapsid (anti-N) antibody levels using World Health Organization standardized binding antibody units per milliliter (BAU/mL). Levels were compared across categorical primary COVID-19 vaccine series received and binary COVID-19 mRNA booster status. The association between anti-S levels and time since most recent vaccination dose was estimated by mixed-effects linear regression. RESULTS In total, 356 students participated, of whom 219 (61.5%) had received a primary vaccine series of Pfizer-BioNTech or Moderna mRNA vaccines and 85 (23.9%) had received vaccines from Sinovac or Sinopharm. Median anti-S levels were significantly higher for mRNA primary vaccine series recipients (2.90 and 2.86 log [BAU/mL], respectively), compared with those who received Sinopharm or Sinovac vaccines (1.63 and 1.95 log [BAU/mL], respectively). Sinopharm and Sinovac vaccine recipients were associated with a significantly faster anti-S decline over time, compared with mRNA vaccine recipients (P <.001). By December, 48/172 (27.9%) participants reported receiving an mRNA COVID-19 vaccine booster, which reduced the anti-S antibody discrepancies between primary series vaccine types. CONCLUSIONS Our work supports the benefit of heterologous boosting against COVID-19. COVID-19 mRNA vaccine booster doses were associated with increases in anti-SARS-CoV-2 antibody levels; following an mRNA booster dose, students with both mRNA and non-mRNA primary series receipt were associated with comparable levels of anti-S IgG.
Collapse
Affiliation(s)
- Peter M DeJonge
- Epidemic Intelligence Service, CDC, Atlanta, Georgia, 30329, USA.
- Wisconsin Department of Health Services, Division of Public Health, Madison, Wisconsin, 53703, USA.
| | | | - Hannah E Segaloff
- Epidemic Intelligence Service, CDC, Atlanta, Georgia, 30329, USA
- Wisconsin Department of Health Services, Division of Public Health, Madison, Wisconsin, 53703, USA
| | - Allen Bateman
- Wisconsin State Laboratory of Hygiene, Madison, Wisconsin, 53703, USA
| | - Alana Sterkel
- Wisconsin State Laboratory of Hygiene, Madison, Wisconsin, 53703, USA
| | - Carol Griggs
- University Health Services, University of Wisconsin - Madison, Madison, Wisconsin, 53703, USA
| | - Jake Baggott
- University Health Services, University of Wisconsin - Madison, Madison, Wisconsin, 53703, USA
| | - Patrick Kelly
- University Health Services, University of Wisconsin - Madison, Madison, Wisconsin, 53703, USA
| | | | | | | | - Glen Abedi
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | | | - Jasmine Y Nakayama
- Epidemic Intelligence Service, CDC, Atlanta, Georgia, 30329, USA
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | - Jasmine Ruffin
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | | | - Almea Matanock
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | | | - Melissa Whaley
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | | | - Kyle DeGruy
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | - Michele Daly
- CDC COVID-19 Response Team, Atlanta, Georgia, 30329, USA
| | - Ryan Westergaard
- Wisconsin Department of Health Services, Division of Public Health, Madison, Wisconsin, 53703, USA
| | | | | |
Collapse
|
84
|
Atef S, Al Hosani F, AbdelWareth L, Al-Rifai RH, Abuyadek R, Jabari A, Ali R, Altrabulsi B, Dunachie S, Alatoom A, Donnelly JG. Susceptibility to reinfection with SARS-CoV-2 virus relative to existing antibody concentrations and T cell response. Int J Infect Dis 2023; 131:100-110. [PMID: 36702370 PMCID: PMC9870609 DOI: 10.1016/j.ijid.2023.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVES We investigated the reinfection rate of vaccinated or convalescent immunized SARS-CoV-2 in 952 expatriate workers with SARS-CoV-2 serological antibody (Ab) patterns and surrogate T cell memory at recruitment and follow-up. METHODS Trimeric spike, nucleocapsid, and neutralizing Abs were measured, along with a T cell stimulation assay, targeting SARS-CoV-2 memory in clusters of differentiation (CD) 4+ and CD8+ T cells. The subjects were then followed up for reinfection for up to 6 months. RESULTS The seroprevalence positivity at enrollment was greater than 99%. The T cell reactivity in this population was 38.2%. Of the 149 (15.9%) participants that were reinfected during the follow-up period (74.3%) had nonreactive T cells at enrollment. Those who had greater than 100 binding Ab units/ml increase from the median concentration of antispike immunoglobulin G Abs had a 6% reduction in the risk of infection. Those who were below the median concentration had a 78% greater risk of infection. CONCLUSION Significant immune protection from reinfection was observed in those who retained T cell activation memory. Additional protection was observed when the antispike was greater than the median value.
Collapse
Affiliation(s)
- Shereen Atef
- National Reference Laboratory, Abu Dhabi, United Arab Emirates; Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | | | - Laila AbdelWareth
- National Reference Laboratory, Abu Dhabi, United Arab Emirates; Cleveland Clinic, Abu Dhabi, United Arab Emirates
| | - Rami H Al-Rifai
- Institute of Public Health, College of Medicine and Health Sciences, Abu Dhabi, United Arab Emirates
| | - Rowan Abuyadek
- Abu Dhabi Public Health Center, Abu Dhabi, United Arab Emirates; High Institute of Public Health, Alexandria University, Alexandria, Egypt
| | - Andrea Jabari
- New-York University, Abu Dhabi, United Arab Emirates
| | - Raghib Ali
- New-York University, Abu Dhabi, United Arab Emirates; MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Basel Altrabulsi
- National Reference Laboratory, Abu Dhabi, United Arab Emirates; Cleveland Clinic, Abu Dhabi, United Arab Emirates
| | | | - Adnan Alatoom
- National Reference Laboratory, Abu Dhabi, United Arab Emirates; Cleveland Clinic, Abu Dhabi, United Arab Emirates
| | | |
Collapse
|
85
|
Seya T, Shingai M, Kawakita T, Matsumoto M. Two Modes of Th1 Polarization Induced by Dendritic-Cell-Priming Adjuvant in Vaccination. Cells 2023; 12:1504. [PMID: 37296625 PMCID: PMC10252737 DOI: 10.3390/cells12111504] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Viral infections are usually accompanied by systemic cytokinemia. Vaccines need not necessarily mimic infection by inducing cytokinemia, but must induce antiviral-acquired immunity. Virus-derived nucleic acids are potential immune-enhancers and particularly good candidates as adjuvants in vaccines in mouse models. The most important nucleic-acid-sensing process involves the dendritic cell (DC) Toll-like receptor (TLR), which participates in the pattern recognition of foreign DNA/RNA structures. Human CD141+ DCs preferentially express TLR3 in endosomes and recognize double-stranded RNA. Antigen cross-presentation occurs preferentially in this subset of DCs (cDCs) via the TLR3-TICAM-1-IRF3 axis. Another subset, plasmacytoid DCs (pDCs), specifically expresses TLR7/9 in endosomes. They then recruit the MyD88 adaptor, and potently induce type I interferon (IFN-I) and proinflammatory cytokines to eliminate the virus. Notably, this inflammation leads to the secondary activation of antigen-presenting cDCs. Hence, the activation of cDCs via nucleic acids involves two modes: (i) with bystander effect of inflammation and (ii) without inflammation. In either case, the acquired immune response finally occurs with Th1 polarity. The level of inflammation and adverse events depend on the TLR repertoire and the mode of response to their agonists in the relevant DC subsets, and could be predicted by assessing the levels of cytokines/chemokines and T cell proliferation in vaccinated subjects. The main differences in the mode of vaccine sought in infectious diseases and cancer are defined by whether it is prophylactic or therapeutic, whether it can deliver sufficient antigens to cDCs, and how it behaves in the microenvironment of the lesion. Adjuvant can be selected on a case-to-case basis.
Collapse
Affiliation(s)
- Tsukasa Seya
- Nebuta Research Institute for Life Sciences, Aomori University, Aomori 030-0943, Japan;
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
- Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (M.S.); (T.K.)
| | - Masashi Shingai
- Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (M.S.); (T.K.)
- Division of Biologics Development, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan
- International Collaboration Unit, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo 001-0021, Japan
| | - Tomomi Kawakita
- Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (M.S.); (T.K.)
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo 001-0021, Japan
| | - Misako Matsumoto
- Nebuta Research Institute for Life Sciences, Aomori University, Aomori 030-0943, Japan;
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
- Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (M.S.); (T.K.)
| |
Collapse
|
86
|
Wei J, Matthews PC, Stoesser N, Newton JN, Diamond I, Studley R, Taylor N, Bell JI, Farrar J, Kolenchery J, Marsden BD, Hoosdally S, Jones EY, Stuart DI, Crook DW, Peto TEA, Walker AS, Pouwels KB, Eyre DW. Protection against SARS-CoV-2 Omicron BA.4/5 variant following booster vaccination or breakthrough infection in the UK. Nat Commun 2023; 14:2799. [PMID: 37193713 PMCID: PMC10187514 DOI: 10.1038/s41467-023-38275-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/21/2023] [Indexed: 05/18/2023] Open
Abstract
Following primary SARS-CoV-2 vaccination, whether boosters or breakthrough infections provide greater protection against SARS-CoV-2 infection is incompletely understood. Here we investigated SARS-CoV-2 antibody correlates of protection against new Omicron BA.4/5 (re-)infections and anti-spike IgG antibody trajectories after a third/booster vaccination or breakthrough infection following second vaccination in 154,149 adults ≥18 y from the United Kingdom general population. Higher antibody levels were associated with increased protection against Omicron BA.4/5 infection and breakthrough infections were associated with higher levels of protection at any given antibody level than boosters. Breakthrough infections generated similar antibody levels to boosters, and the subsequent antibody declines were slightly slower than after boosters. Together our findings show breakthrough infection provides longer-lasting protection against further infections than booster vaccinations. Our findings, considered alongside the risks of severe infection and long-term consequences of infection, have important implications for vaccine policy.
Collapse
Affiliation(s)
- Jia Wei
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Philippa C Matthews
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- The Francis Crick Institute, 1 Midland Road, London, UK
- Division of infection and immunity, University College London, London, UK
| | - Nicole Stoesser
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - John N Newton
- European Centre for Environment and Human Health, University of Exeter, Truro, UK
| | | | | | | | - John I Bell
- Office of the Regius Professor of Medicine, University of Oxford, Oxford, UK
| | | | - Jaison Kolenchery
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Brian D Marsden
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Sarah Hoosdally
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - E Yvonne Jones
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - David I Stuart
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Derrick W Crook
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Tim E A Peto
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - A Sarah Walker
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- MRC Clinical Trials Unit at UCL, UCL, London, UK
| | - Koen B Pouwels
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- Health Economics Research Centre, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - David W Eyre
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK.
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK.
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
| |
Collapse
|
87
|
Prather AA, Dutcher EG, Robinson J, Lin J, Blackburn E, Hecht FM, Mason AE, Fromer E, Merino B, Frazier R, O'Bryan J, Drury S, Epel ES. Predictors of long-term neutralizing antibody titers following COVID-19 vaccination by three vaccine types: the BOOST study. Sci Rep 2023; 13:6505. [PMID: 37160978 PMCID: PMC10170073 DOI: 10.1038/s41598-023-33320-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/11/2023] [Indexed: 05/11/2023] Open
Abstract
As concerns related to the COVID-19 pandemic continue, it is critical to understand the impact of vaccination type on neutralizing antibody response durability as well as to identify individual difference factors related to decline in neutralization. This was a head-to-head comparison study following 498 healthy, community volunteers who received the BNT162b2 (n = 287), mRNA-1273 (n = 149), and Ad26.COV2.S (n = 62). Participants completed questionnaires and underwent blood draws prior to vaccination, 1 month, and 6 months after the vaccination series, and neutralizing antibody (nAB) titers at 1- and 6-months post vaccination were quantified using a high-throughput pseudovirus assay. Over 6 months of follow-up, nABs declined in recipients of BNT162b2 and mRNA-1273, while nABs in recipients of Ad26.COV2.S showed a significant increase. At the 6-month time point, nABs to Ad26.COV2.S were significantly higher than nABs to BNT162b2 and equivalent to mRNA-1273. Irrespective of follow-up timing, being older was associated with lower nAB for participants who received BNT162b2 and Ad26.COV2.S but not for those who received mRNA-1273. A higher baseline BMI was associated with a lower nAB for Ad26.COV2.S recipients but not for recipients of other vaccines. Women and non-smokers showed higher nAB compared to men and current smokers, respectively. The durability of neutralizing antibody responses differed by vaccine type and several sociodemographic factors that predicted response. These findings may inform booster recommendations in the future.
Collapse
Affiliation(s)
- Aric A Prather
- Center for Health and Community, University of California, 675 18th St., San Francisco, CA, 94107, USA.
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, USA.
| | - Ethan G Dutcher
- Center for Health and Community, University of California, 675 18th St., San Francisco, CA, 94107, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, USA
| | - James Robinson
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, USA
| | - Jue Lin
- Department of Biochemistry and Biophysics, University of California, San Francisco, USA
| | - Elizabeth Blackburn
- Department of Biochemistry and Biophysics, University of California, San Francisco, USA
| | - Frederick M Hecht
- Department of Medicine, University of California, San Francisco, USA
- Osher Center for Integrative Health, University of California, San Francisco, USA
| | - Ashley E Mason
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, USA
- Osher Center for Integrative Health, University of California, San Francisco, USA
| | - Elena Fromer
- Center for Health and Community, University of California, 675 18th St., San Francisco, CA, 94107, USA
| | - Bresh Merino
- Center for Health and Community, University of California, 675 18th St., San Francisco, CA, 94107, USA
| | - Remi Frazier
- Academic Research Systems, University of California, San Francisco, USA
| | - Julia O'Bryan
- Center for Health and Community, University of California, 675 18th St., San Francisco, CA, 94107, USA
| | - Stacy Drury
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, USA
- Department of Psychiatry, Tulane University School of Medicine, New Orleans, USA
| | - Elissa S Epel
- Center for Health and Community, University of California, 675 18th St., San Francisco, CA, 94107, USA.
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, USA.
| |
Collapse
|
88
|
Espíndola OM, Fuller TL, de Araújo MF, Tort LFL, Guaraldo L, Calvet G, Resende P, Bonaldo M, Whitworth J, Smith C, Siqueira M, Brasil P. Reduced ability to neutralize the Omicron variant among adults after infection and complete vaccination with BNT162b2, ChAdOx1, or CoronaVac and heterologous boosting. Sci Rep 2023; 13:7437. [PMID: 37156846 PMCID: PMC10165291 DOI: 10.1038/s41598-023-34035-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/22/2023] [Indexed: 05/10/2023] Open
Abstract
COVID-19 vaccines have dramatically reduced rates of severe infection requiring hospitalization. However, SARS-CoV-2 variants have reduced vaccine effectiveness at preventing any symptomatic infection. This real-world study analyzed binding and neutralizing antibodies generated after complete vaccination and boosting across three vaccine platforms. Binding antibodies decayed most slowly in people under 60 with hybrid immunity. Neutralizing antibodies against Omicron BA.1 were reduced compared to other variants. The anamnestic anti-spike IgG response to the first boost was more pronounced than after the second boost. Monitoring of the effects of SARS-CoV-2 mutations on disease severity and the effectiveness of therapeutics is warranted.
Collapse
Affiliation(s)
- Otávio Melo Espíndola
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (Fiocruz), Avenida Brasil 4365, Rio de Janeiro, RJ, 21040-900, Brazil.
| | - Trevon L Fuller
- Institute of the Environment and Sustainability, University of California Los Angeles, Los Angeles, 90095, USA.
| | - Mia Ferreira de Araújo
- Laboratory of Respiratory Viruses and Measles National Influenza Centre (GISRS-WHO)-Americas Regional Reference Lab for Measles and Rubella-Reference Laboratory for COVID-19 (WHO), Oswaldo Cruz Institute (IOC), Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Luis Fernando Lopez Tort
- Laboratory of Respiratory Viruses and Measles National Influenza Centre (GISRS-WHO)-Americas Regional Reference Lab for Measles and Rubella-Reference Laboratory for COVID-19 (WHO), Oswaldo Cruz Institute (IOC), Fiocruz, Rio de Janeiro, 21040-900, Brazil
- Laboratory of Molecular Virology, Biological Sciences Department, Universidad de la República, Salto, Uruguay
| | - Lusiele Guaraldo
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (Fiocruz), Avenida Brasil 4365, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Guilherme Calvet
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (Fiocruz), Avenida Brasil 4365, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Paola Resende
- Laboratory of Respiratory Viruses and Measles National Influenza Centre (GISRS-WHO)-Americas Regional Reference Lab for Measles and Rubella-Reference Laboratory for COVID-19 (WHO), Oswaldo Cruz Institute (IOC), Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Myrna Bonaldo
- Laboratory of Molecular Biology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Jimmy Whitworth
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Chris Smith
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Marilda Siqueira
- Laboratory of Respiratory Viruses and Measles National Influenza Centre (GISRS-WHO)-Americas Regional Reference Lab for Measles and Rubella-Reference Laboratory for COVID-19 (WHO), Oswaldo Cruz Institute (IOC), Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Patrícia Brasil
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (Fiocruz), Avenida Brasil 4365, Rio de Janeiro, RJ, 21040-900, Brazil.
| |
Collapse
|
89
|
Zhao D, Chen X, Wang L, Zhang J, Zhao Z, Yue N, Zhu Y, Fei W, Li X, Tan L, He W. Bidirectional and persistent immunomodulation of Astragalus polysaccharide as an adjuvant of influenza and recombinant SARS-CoV-2 vaccine. Int J Biol Macromol 2023; 234:123635. [PMID: 36801224 PMCID: PMC9932796 DOI: 10.1016/j.ijbiomac.2023.123635] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023]
Abstract
Respiratory viral infections, such as coronavirus disease of 2019 (COVID-19) and influenza, cause significant morbidity and mortality and have become a worldwide public health concern with tremendous economic and societal burdens. Vaccination is a major strategy for preventing infections. However, some new vaccines have an unmet need for impairing responses in certain individuals, especially COVID-19 vaccines, despite ongoing vaccine and adjuvant research. Here, we evaluated the effectiveness of Astragalus polysaccharide (APS), a bioactive polysaccharide extracted from the traditional Chinese herb Astragalus membranaceus as an immune adjuvant to regulate the efficacy of influenza split vaccine (ISV) and recombinant severe acute respiratory syndrome (SARS)-Cov-2 vaccine in mice. Our data indicated that APS as an adjuvant can facilitate the induction of high levels of hemagglutination inhibition (HAI) titer and specific antibody immunoglobulin G (IgG) and confer protection against the lethal challenge of influenza A viruses, including increased survival and amelioration of weight loss in mice immunized with the ISV. RNA sequencing (RNA-seq) analysis revealed that the NF-κB and Fc gamma R-mediated phagocytosis signaling pathways are essential for the immune response of mice immunized with the recombinant SARS-Cov-2 vaccine (RSV). Another important finding was that bidirectional immunomodulation of APS on cellular and humoral immunity was observed, and APS-adjuvant-induced antibodies persisted at a high level for at least 20 weeks. These findings suggest that APS is a potent adjuvant for influenza and COVID-19 vaccines, and has the advantages of bidirectional immunoregulation and persistent immunity.
Collapse
Affiliation(s)
- Danping Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiuhong Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Linyuan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| | - Jianjun Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Zhongpeng Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| | - Na Yue
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yingli Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wenting Fei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyu Li
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lingyun Tan
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Wei He
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
90
|
Nafilyan V, Bermingham CR, Ward IL, Morgan J, Zaccardi F, Khunti K, Stanborough J, Banerjee A, Doidge JC. Risk of death following COVID-19 vaccination or positive SARS-CoV-2 test in young people in England. Nat Commun 2023; 14:1541. [PMID: 36973247 PMCID: PMC10043280 DOI: 10.1038/s41467-023-36494-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 02/02/2023] [Indexed: 03/29/2023] Open
Abstract
Several studies have reported associations between COVID-19 vaccination and risk of cardiac diseases, especially in young people; the impact on mortality, however, remains unclear. We use national, linked electronic health data in England to assess the impact of COVID-19 vaccination and positive SARS-CoV-2 tests on the risk of cardiac and all-cause mortality in young people (12 to 29 years) using a self-controlled case series design. Here, we show there is no significant increase in cardiac or all-cause mortality in the 12 weeks following COVID-19 vaccination compared to more than 12 weeks after any dose. However, we find an increase in cardiac death in women after a first dose of non mRNA vaccines. A positive SARS-CoV-2 test is associated with increased cardiac and all-cause mortality among people vaccinated or unvaccinated at time of testing.
Collapse
Affiliation(s)
- Vahé Nafilyan
- Data and Analysis for Social Care and Health, Office for National Statistics, Newport, NP10 8XG, UK.
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK.
| | - Charlotte R Bermingham
- Data and Analysis for Social Care and Health, Office for National Statistics, Newport, NP10 8XG, UK.
| | - Isobel L Ward
- Data and Analysis for Social Care and Health, Office for National Statistics, Newport, NP10 8XG, UK
| | - Jasper Morgan
- Data and Analysis for Social Care and Health, Office for National Statistics, Newport, NP10 8XG, UK
| | - Francesco Zaccardi
- Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Kamlesh Khunti
- Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Julie Stanborough
- Data and Analysis for Social Care and Health, Office for National Statistics, Newport, NP10 8XG, UK
| | - Amitava Banerjee
- Institute of Health Informatics, University College London, London, NW1 2DA, UK
| | - James C Doidge
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
- Intensive Care National Audit and Research Centre, London, UK
| |
Collapse
|
91
|
Yoon J, Park B, Kim H, Choi S, Jung D. Korean Red Ginseng Potentially Improves Maintaining Antibodies after COVID-19 Vaccination: A 24-Week Longitudinal Study. Nutrients 2023; 15:nu15071584. [PMID: 37049424 PMCID: PMC10097014 DOI: 10.3390/nu15071584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Despite the effectiveness and safety of COVID-19 vaccines, vaccine-induced responses decline over time; thus, booster vaccines have been approved globally. In addition, interest in natural compounds capable of improving host immunity has increased. This study aimed to examine the effect of Korean Red Ginseng (KRG) on virus-specific antibodies after COVID-19 vaccination. We conducted a 24 week clinical pilot study of 350 healthy subjects who received two doses of the COVID-19 vaccine and a booster vaccination (third dose). These subjects were randomized 1:2 to the KRG and control groups. We evaluated antibody response five times: just before the second dose (baseline), 2 weeks, 4 weeks, 12 weeks after the second dose, and 4 weeks after the third dose. The primary endpoints were changes in COVID-19 spike antibody titers and neutralizing antibody titers. The antibody formation rate of the KRG group was sustained higher than that of the control group for 12 weeks after the second dose. This trend was prominently observed in those above 50 years old. We found that KRG can help to increase and maintain vaccine response, highlighting that KRG could potentially be used as an immunomodulator with COVID-19 vaccines.
Collapse
Affiliation(s)
- Jihyun Yoon
- Department of Family Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Byoungjin Park
- Department of Family Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Heejung Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seungjun Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Laboratory Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, Republic of Korea
| | - Donghyuk Jung
- Department of Family Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
92
|
Marking U, Havervall S, Norin NG, Bladh O, Christ W, Gordon M, Ng H, Blom K, Phillipson M, Mangsbo S, Alm JJ, Smed-Sörensen A, Nilsson P, Hober S, Åberg M, Klingström J, Thålin C. Correlates of protection and viral load trajectories in omicron breakthrough infections in triple vaccinated healthcare workers. Nat Commun 2023; 14:1577. [PMID: 36949041 PMCID: PMC10031702 DOI: 10.1038/s41467-023-36984-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 02/27/2023] [Indexed: 03/24/2023] Open
Abstract
Vaccination offers protection against severe COVID-19 caused by SARS-CoV-2 omicron but is less effective against infection. Characteristics such as serum antibody titer correlation to protection, viral abundance and clearance of omicron infection in vaccinated individuals are scarce. We present a 4-week twice-weekly SARS-CoV-2 qPCR screening in 368 triple vaccinated healthcare workers. Spike-specific IgG levels, neutralization titers and mucosal spike-specific IgA-levels were determined at study start and qPCR-positive participants were sampled repeatedly for two weeks. 81 (cumulative incidence 22%) BA.1, BA.1.1 and BA.2 infections were detected. High serum antibody titers are shown to be protective against infection (p < 0.01), linked to reduced viral load (p < 0.01) and time to viral clearance (p < 0.05). Pre-omicron SARS-CoV-2 infection is independently associated to increased protection against omicron, largely mediated by mucosal spike specific IgA responses (nested models lr test p = 0.02 and 0.008). Only 10% of infected participants remain asymptomatic through the course of their infection. We demonstrate that high levels of vaccine-induced spike-specific WT antibodies are linked to increased protection against infection and to reduced viral load if infected, and suggest that the additional protection offered by pre-omicron SARS-CoV-2 infection largely is mediated by mucosal spike-specific IgA.
Collapse
Affiliation(s)
- Ulrika Marking
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Sebastian Havervall
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Nina Greilert Norin
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Oscar Bladh
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Wanda Christ
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Max Gordon
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Henry Ng
- Department of Medical Cell Biology and SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Kim Blom
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Mia Phillipson
- Department of Medical Cell Biology and SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Sara Mangsbo
- Department of Pharmacy and SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Jessica J Alm
- Department of Microbiology, Tumor and Cell Biology & National Pandemic Center, Karolinska Institutet, Solna, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Nilsson
- Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Sophia Hober
- Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry and SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Jonas Klingström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Charlotte Thålin
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden.
| |
Collapse
|
93
|
Moore SC, Kronsteiner B, Longet S, Adele S, Deeks AS, Liu C, Dejnirattisai W, Reyes LS, Meardon N, Faustini S, Al-Taei S, Tipton T, Hering LM, Angyal A, Brown R, Nicols AR, Dobson SL, Supasa P, Tuekprakhon A, Cross A, Tyerman JK, Hornsby H, Grouneva I, Plowright M, Zhang P, Newman TAH, Nell JM, Abraham P, Ali M, Malone T, Neale I, Phillips E, Wilson JD, Murray SM, Zewdie M, Shields A, Horner EC, Booth LH, Stafford L, Bibi S, Wootton DG, Mentzer AJ, Conlon CP, Jeffery K, Matthews PC, Pollard AJ, Brown A, Rowland-Jones SL, Mongkolsapaya J, Payne RP, Dold C, Lambe T, Thaventhiran JED, Screaton G, Barnes E, Hopkins S, Hall V, Duncan CJA, Richter A, Carroll M, de Silva TI, Klenerman P, Dunachie S, Turtle L. Evolution of long-term vaccine-induced and hybrid immunity in healthcare workers after different COVID-19 vaccine regimens. MED 2023; 4:191-215.e9. [PMID: 36863347 PMCID: PMC9933851 DOI: 10.1016/j.medj.2023.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023]
Abstract
BACKGROUND Both infection and vaccination, alone or in combination, generate antibody and T cell responses against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the maintenance of such responses-and hence protection from disease-requires careful characterization. In a large prospective study of UK healthcare workers (HCWs) (Protective Immunity from T Cells in Healthcare Workers [PITCH], within the larger SARS-CoV-2 Immunity and Reinfection Evaluation [SIREN] study), we previously observed that prior infection strongly affected subsequent cellular and humoral immunity induced after long and short dosing intervals of BNT162b2 (Pfizer/BioNTech) vaccination. METHODS Here, we report longer follow-up of 684 HCWs in this cohort over 6-9 months following two doses of BNT162b2 or AZD1222 (Oxford/AstraZeneca) vaccination and up to 6 months following a subsequent mRNA booster vaccination. FINDINGS We make three observations: first, the dynamics of humoral and cellular responses differ; binding and neutralizing antibodies declined, whereas T and memory B cell responses were maintained after the second vaccine dose. Second, vaccine boosting restored immunoglobulin (Ig) G levels; broadened neutralizing activity against variants of concern, including Omicron BA.1, BA.2, and BA.5; and boosted T cell responses above the 6-month level after dose 2. Third, prior infection maintained its impact driving larger and broader T cell responses compared with never-infected people, a feature maintained until 6 months after the third dose. CONCLUSIONS Broadly cross-reactive T cell responses are well maintained over time-especially in those with combined vaccine and infection-induced immunity ("hybrid" immunity)-and may contribute to continued protection against severe disease. FUNDING Department for Health and Social Care, Medical Research Council.
Collapse
Affiliation(s)
- Shona C Moore
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Stephanie Longet
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sandra Adele
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Alexandra S Deeks
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Chang Liu
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Wanwisa Dejnirattisai
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Division of Emerging Infectious Disease, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Laura Silva Reyes
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Naomi Meardon
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Sian Faustini
- Institute for Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK
| | - Saly Al-Taei
- Institute for Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK
| | - Tom Tipton
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Luisa M Hering
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Adrienn Angyal
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Rebecca Brown
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Alexander R Nicols
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle, UK
| | - Susan L Dobson
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Piyada Supasa
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Aekkachai Tuekprakhon
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew Cross
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Jessica K Tyerman
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle, UK
| | - Hailey Hornsby
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Irina Grouneva
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Megan Plowright
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Peijun Zhang
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Thomas A H Newman
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Jeremy M Nell
- Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Priyanka Abraham
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Mohammad Ali
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Tom Malone
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Isabel Neale
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Eloise Phillips
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Joseph D Wilson
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Oxford University Medical School, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Sam M Murray
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Martha Zewdie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Adrian Shields
- Institute for Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK; University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Emily C Horner
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Lucy H Booth
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Lizzie Stafford
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Daniel G Wootton
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK; Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK; Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Alexander J Mentzer
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Christopher P Conlon
- Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Katie Jeffery
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Philippa C Matthews
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; The Francis Crick Institute, London, UK; Division of Infection and Immunity, University College London, London, UK; Department of Infectious Diseases, University College London Hospital NHS Foundation Trust, London, UK
| | - Andrew J Pollard
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Anthony Brown
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Sarah L Rowland-Jones
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Juthathip Mongkolsapaya
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Rebecca P Payne
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle, UK
| | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | | | - Gavin Screaton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Susan Hopkins
- UK Health Security Agency, London, UK; Faculty of Medicine, Department of Infectious Disease, Imperial College London, London, UK; NIHR Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, University of Oxford, Oxford, UK
| | - Victoria Hall
- UK Health Security Agency, London, UK; NIHR Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, University of Oxford, Oxford, UK
| | - Christopher J A Duncan
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle, UK; Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Alex Richter
- Institute for Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK; University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Miles Carroll
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Thushan I de Silva
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Translational Gastroenterology Unit, University of Oxford, Oxford, UK.
| | - Susanna Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| | - Lance Turtle
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK; Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
94
|
Chibwana MG, Thole HW, Anscombe C, Ashton PM, Green E, Barnes KG, Cornick J, Turner A, Witte D, Nthala S, Thom C, Kanyandula F, Ainani A, Mtike N, Tambala H, N’goma V, Mwafulirwa D, Asima E, Morton B, Gmeiner M, Gundah Z, Kawalazira G, French N, Feasey N, Heyderman RS, Swarthout TD, Jambo KC. Different clinical features in Malawian outpatients presenting with COVID-19 prior to and during Omicron variant dominance: A prospective observational study. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0001575. [PMID: 36963090 PMCID: PMC10022204 DOI: 10.1371/journal.pgph.0001575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/03/2023] [Indexed: 03/11/2023]
Abstract
The SARS-CoV-2 Omicron variant has resulted in a high number of cases, but a relatively low incidence of severe disease and deaths, compared to the pre-Omicron variants. Therefore, we assessed the differences in symptom prevalence between Omicron and pre-Omicron infections in a sub-Saharan African population. We collected data from outpatients presenting at two primary healthcare facilities in Blantyre, Malawi, from November 2020 to March 2022. Eligible participants were aged >1month old, with signs suggestive of COVID-19, and those not suspected of COVID-19, from whom we collected nasopharyngeal swabs for SARS-CoV-2 PCR testing, and sequenced positive samples to identify infecting-variants. In addition, we calculated the risk of presenting with a given symptom in individuals testing SARS-CoV-2 PCR positive before and during the Omicron variant-dominated period. Among 5176 participants, 6.4% were under 5, and 77% were aged 18 to 50 years. SARS-CoV-2 infection prevalence peaked in January 2021 (Beta), July 2021 (Delta), and December 2021 (Omicron). We found that cough (risk ratio (RR), 1.50; 95% confidence interval (CI), 1.00 to 2.30), fatigue (RR 2.27; 95% CI, 1.29 to 3.86) and headache (RR 1.64; 95% CI, 1.15 to 2.34) were associated with a high risk of SARS-CoV-2 infection during the pre-Omicron period. In comparison, only headache (RR 1.41; 95% CI, 1.07 to 1.86) did associate with a high risk of SARS-CoV-2 infection during the Omicron-dominated period. In conclusion, clinical symptoms associated with Omicron infection differed from prior variants and were harder to identify clinically with current symptom guidelines. Our findings encourage regular review of case definitions and testing policies to ensure case ascertainment.
Collapse
Affiliation(s)
| | | | - Cat Anscombe
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Philip M. Ashton
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Edward Green
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Kayla G. Barnes
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Harvard School of Public Health, Boston, MA, United States of America
- Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
- University of Glasgow MRC Centre for Virus Research, Glasgow, United Kingdom
| | - Jen Cornick
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Ann Turner
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Desiree Witte
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Sharon Nthala
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
| | - Chikondi Thom
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
| | | | - Anna Ainani
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
| | - Natasha Mtike
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
| | - Hope Tambala
- Blantyre District Health Office, Ministry of Health, Blantyre, Malawi
| | | | | | - Erick Asima
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
| | - Ben Morton
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Markus Gmeiner
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
| | - Zaziwe Gundah
- Blantyre District Health Office, Ministry of Health, Blantyre, Malawi
| | - Gift Kawalazira
- Blantyre District Health Office, Ministry of Health, Blantyre, Malawi
| | - Neil French
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Nicholas Feasey
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Robert S. Heyderman
- NIHR Global Health Research Unit on Mucosal Pathogens, University College London, London, United Kingdom
| | - Todd D. Swarthout
- NIHR Global Health Research Unit on Mucosal Pathogens, University College London, London, United Kingdom
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Kondwani C. Jambo
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Kamuzu University of Health Sciences, Blantyre, Malawi
| |
Collapse
|
95
|
Cai JP, Luo C, Wang K, Cao H, Chen LL, Zhang X, Han Y, Yin F, Zhang AJ, Chu H, Yuan S, Kok KH, To KKW, Chen H, Chen Z, Jin DY, Yuen KY, Chan JFW. Intranasal Boosting with Spike Fc-RBD of Wild-Type SARS-CoV-2 Induces Neutralizing Antibodies against Omicron Subvariants and Reduces Viral Load in the Nasal Turbinate of Mice. Viruses 2023; 15:687. [PMID: 36992395 PMCID: PMC10052291 DOI: 10.3390/v15030687] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/27/2023] [Accepted: 03/04/2023] [Indexed: 03/08/2023] Open
Abstract
The emergence of new immune-evasive severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants and subvariants outpaces the development of vaccines specific against the dominant circulating strains. In terms of the only accepted immune correlate of protection, the inactivated whole-virion vaccine using wild-type SARS-CoV-2 spike induces a much lower serum neutralizing antibody titre against the Omicron subvariants. Since the inactivated vaccine given intramuscularly is one of the most commonly used coronavirus disease 2019 (COVID-19) vaccines in developing regions, we tested the hypothesis that intranasal boosting after intramuscular priming would provide a broader level of protection. Here, we showed that one or two intranasal boosts with the Fc-linked trimeric spike receptor-binding domain from wild-type SARS-CoV-2 can induce significantly higher serum neutralizing antibodies against wild-type SARS-CoV-2 and the Omicron subvariants, including BA.5.2 and XBB.1, with a lower titre in the bronchoalveolar lavage of vaccinated Balb/c mice than vaccination with four intramuscular doses of inactivated whole virion vaccine. The intranasally vaccinated K18-hACE2-transgenic mice also had a significantly lower nasal turbinate viral load, suggesting a better protection of the upper airway, which is the predilected site of infection by Omicron subvariants. This intramuscular priming and intranasal boosting approach that achieves broader cross-protection against Omicron variants and subvariants may lengthen the interval required for changing the vaccine immunogen from months to years.
Collapse
Affiliation(s)
- Jian-Piao Cai
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Cuiting Luo
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kun Wang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Hehe Cao
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Lin-Lei Chen
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Xiaojuan Zhang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yuting Han
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Feifei Yin
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou 570100, China
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou 570100, China
| | - Anna Jinxia Zhang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Kin-Hang Kok
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Kelvin Kai-Wang To
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
- Guangzhou Laboratory, Guangdong Province, Guangzhou 510000, China
| | - Honglin Chen
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
- Guangzhou Laboratory, Guangdong Province, Guangzhou 510000, China
| | - Zhiwei Chen
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
- Guangzhou Laboratory, Guangdong Province, Guangzhou 510000, China
| | - Dong-Yan Jin
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
- Guangzhou Laboratory, Guangdong Province, Guangzhou 510000, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou 570100, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
- Guangzhou Laboratory, Guangdong Province, Guangzhou 510000, China
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou 570100, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
- Guangzhou Laboratory, Guangdong Province, Guangzhou 510000, China
| |
Collapse
|
96
|
Li J, Huang F, Ma Q, Guo W, Feng K, Huang T, Cai YD. Identification of genes related to immune enhancement caused by heterologous ChAdOx1-BNT162b2 vaccines in lymphocytes at single-cell resolution with machine learning methods. Front Immunol 2023; 14:1131051. [PMID: 36936955 PMCID: PMC10017451 DOI: 10.3389/fimmu.2023.1131051] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
The widely used ChAdOx1 nCoV-19 (ChAd) vector and BNT162b2 (BNT) mRNA vaccines have been shown to induce robust immune responses. Recent studies demonstrated that the immune responses of people who received one dose of ChAdOx1 and one dose of BNT were better than those of people who received vaccines with two homologous ChAdOx1 or two BNT doses. However, how heterologous vaccines function has not been extensively investigated. In this study, single-cell RNA sequencing data from three classes of samples: volunteers vaccinated with heterologous ChAdOx1-BNT and volunteers vaccinated with homologous ChAd-ChAd and BNT-BNT vaccinations after 7 days were divided into three types of immune cells (3654 B, 8212 CD4+ T, and 5608 CD8+ T cells). To identify differences in gene expression in various cell types induced by vaccines administered through different vaccination strategies, multiple advanced feature selection methods (max-relevance and min-redundancy, Monte Carlo feature selection, least absolute shrinkage and selection operator, light gradient boosting machine, and permutation feature importance) and classification algorithms (decision tree and random forest) were integrated into a computational framework. Feature selection methods were in charge of analyzing the importance of gene features, yielding multiple gene lists. These lists were fed into incremental feature selection, incorporating decision tree and random forest, to extract essential genes, classification rules and build efficient classifiers. Highly ranked genes include PLCG2, whose differential expression is important to the B cell immune pathway and is positively correlated with immune cells, such as CD8+ T cells, and B2M, which is associated with thymic T cell differentiation. This study gave an important contribution to the mechanistic explanation of results showing the stronger immune response of a heterologous ChAdOx1-BNT vaccination schedule than two doses of either BNT or ChAdOx1, offering a theoretical foundation for vaccine modification.
Collapse
Affiliation(s)
- Jing Li
- School of Computer Science, Baicheng Normal University, Baicheng, Jilin, China
| | - FeiMing Huang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - QingLan Ma
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Wei Guo
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - KaiYan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou, China
| | - Tao Huang
- CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Science, Shanghai, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
97
|
Atchison CJ, Moshe M, Brown JC, Whitaker M, Wong NCK, Bharath AA, McKendry RA, Darzi A, Ashby D, Donnelly CA, Riley S, Elliott P, Barclay WS, Cooke GS, Ward H. Validity of Self-testing at Home With Rapid Severe Acute Respiratory Syndrome Coronavirus 2 Antibody Detection by Lateral Flow Immunoassay. Clin Infect Dis 2023; 76:658-666. [PMID: 35913410 PMCID: PMC9384551 DOI: 10.1093/cid/ciac629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/14/2022] [Accepted: 07/28/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND We explore severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibody lateral flow immunoassay (LFIA) performance under field conditions compared to laboratory-based electrochemiluminescence immunoassay (ECLIA) and live virus neutralization. METHODS In July 2021, 3758 participants performed, at home, a self-administered Fortress LFIA on finger-prick blood, reported and submitted a photograph of the result, and provided a self-collected capillary blood sample for assessment of immunoglobulin G (IgG) antibodies using the Roche Elecsys® Anti-SARS-CoV-2 ECLIA. We compared the self-reported LFIA result to the quantitative ECLIA and checked the reading of the LFIA result with an automated image analysis (ALFA). In a subsample of 250 participants, we compared the results to live virus neutralization. RESULTS Almost all participants (3593/3758, 95.6%) had been vaccinated or reported prior infection. Overall, 2777/3758 (73.9%) were positive on self-reported LFIA, 2811/3457 (81.3%) positive by LFIA when ALFA-reported, and 3622/3758 (96.4%) positive on ECLIA (using the manufacturer reference standard threshold for positivity of 0.8 U mL-1). Live virus neutralization was detected in 169 of 250 randomly selected samples (67.6%); 133/169 were positive with self-reported LFIA (sensitivity 78.7%; 95% confidence interval [CI]: 71.8, 84.6), 142/155 (91.6%; 95% CI: 86.1, 95.5) with ALFA, and 169 (100%; 95% CI: 97.8, 100.0) with ECLIA. There were 81 samples with no detectable virus neutralization; 47/81 were negative with self-reported LFIA (specificity 58.0%; 95% CI: 46.5, 68.9), 34/75 (45.3%; 95% CI: 33.8, 57.3) with ALFA, and 0/81 (0%; 95% CI: 0, 4.5) with ECLIA. CONCLUSIONS Self-administered LFIA is less sensitive than a quantitative antibody test, but the positivity in LFIA correlates better than the quantitative ECLIA with virus neutralization.
Collapse
Affiliation(s)
- Christina J Atchison
- School of Public Health, Imperial College London, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Maya Moshe
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Jonathan C Brown
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Matthew Whitaker
- School of Public Health, Imperial College London, London, United Kingdom
| | - Nathan C K Wong
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Anil A Bharath
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Rachel A McKendry
- London Centre for Nanotechnology & Division of Medicine, University College London, London, United Kingdom
- Division of Medicine, University College London, London, United Kingdom
| | - Ara Darzi
- Imperial College Healthcare NHS Trust, London, United Kingdom
- Institute of Global Health Innovation at Imperial College London, London, United Kingdom
| | - Deborah Ashby
- School of Public Health, Imperial College London, London, United Kingdom
| | - Christl A Donnelly
- School of Public Health, Imperial College London, London, United Kingdom
- Department of Statistics, University of Oxford, Oxford, United Kingdom
- MRC Centre for Global infectious Disease Analysis and Abdul Latif Jameel Institute for Disease and Emergency Analytics, Imperial College London, London, United Kingdom
| | - Steven Riley
- School of Public Health, Imperial College London, London, United Kingdom
- MRC Centre for Global infectious Disease Analysis and Abdul Latif Jameel Institute for Disease and Emergency Analytics, Imperial College London, London, United Kingdom
| | - Paul Elliott
- School of Public Health, Imperial College London, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
- National Institute for Health Research Imperial Biomedical Research Centre, London, United Kingdom
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom
- Health Data Research (HDR) UK London at Imperial College, London, United Kingdom
- UK Dementia Research Institute at Imperial College, London, United Kingdom
| | - Wendy S Barclay
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Graham S Cooke
- Imperial College Healthcare NHS Trust, London, United Kingdom
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- National Institute for Health Research Imperial Biomedical Research Centre, London, United Kingdom
| | - Helen Ward
- School of Public Health, Imperial College London, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
- MRC Centre for Global infectious Disease Analysis and Abdul Latif Jameel Institute for Disease and Emergency Analytics, Imperial College London, London, United Kingdom
- National Institute for Health Research Imperial Biomedical Research Centre, London, United Kingdom
| |
Collapse
|
98
|
Mseka UL, Mandolo J, Nyoni K, Divala O, Kambalame D, Mapemba D, Kamzati M, Chibwe I, Henrion MY, Manda K, Thindwa D, Mvula M, Odala B, Kamng'ona R, Dzinza N, Jere KC, Feasey N, Ho A, Amoah AS, Gordon M, Swarthout TD, Crampin A, Heyderman RS, Kagoli M, Chitsa-Banda E, Mitambo C, Phuka J, Chilima B, Kasambara W, Jambo KC, Chauma-Mwale A. Omicron B.1.1.529 variant infections associated with severe disease are uncommon in a COVID-19 under-vaccinated, high SARS-CoV-2 seroprevalence population in Malawi. EClinicalMedicine 2023; 56:101800. [PMID: 36600885 PMCID: PMC9800171 DOI: 10.1016/j.eclinm.2022.101800] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/06/2022] [Accepted: 12/06/2022] [Indexed: 12/31/2022] Open
Abstract
Background The B.1.1.529 (Omicron) variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in the fourth COVID-19 pandemic wave across the southern African region, including Malawi. The seroprevalence of SARS-CoV-2 antibodies and their association with epidemiological trends of hospitalisations and deaths are needed to aid locally relevant public health policy decisions. Methods We conducted a population-based serosurvey from December 27, 2021 to January 17, 2022, in 7 districts across Malawi to determine the seroprevalence of SARS-CoV-2 antibodies. Serum samples were tested for antibodies against SARS-CoV-2 receptor binding domain using WANTAI SARS-CoV-2 Receptor Binding Domain total antibody commercial enzyme-linked immunosorbent assay (ELISA). We also evaluated COVID-19 epidemiologic trends in Malawi, including cases, hospitalisations and deaths from April 1, 2021 through April 30, 2022, collected using the routine national COVID-19 reporting system. A multivariable logistic regression model was developed to investigate the factors associated with SARS-CoV-2 seropositivity. Findings Serum samples were analysed from 4619 participants (57% female; 60% aged 18-50 years), of whom 878/3794 (23%) of vaccine eligible adults had received a single dose of any COVID-19 vaccine. The overall assay-adjusted seroprevalence was 83.7% (95% confidence interval (CI), 79.3%-93.4%). Seroprevalence was lowest among children <13 years of age (66%) and highest among adults 18-50 years of age (82%). Seroprevalence was higher among vaccinated compared to unvaccinated participants (1 dose, 94% vs. 77%, adjusted odds ratio 4.89 [95% CI, 3.43-7.22]; 2 doses, 97% vs. 77%, aOR 6.62 [95% CI, 4.14-11.3]). Urban residents were more likely to be seropositive than those from rural settings (91% vs. 78%, aOR 2.76 [95% CI, 2.16-3.55]). There was at least a two-fold reduction in the proportion of hospitalisations and deaths among the reported cases in the fourth wave compared to the third wave (hospitalisations, 10.7% (95% CI, 10.2-11.3) vs. 4.86% (95% CI, 4.52-5.23), p < 0.0001; deaths, 3.48% (95% CI, 3.18-3.81) vs. 1.15% (95% CI, 1.00-1.34), p < 0.0001). Interpretation We report reduction in proportion of hospitalisations and deaths from SARS-CoV-2 infections during the Omicron variant dominated wave in Malawi, in the context of high SARS-CoV-2 seroprevalence and low COVID-19 vaccination coverage. These findings suggest that COVID-19 vaccination policy in high seroprevalence settings may need to be amended from mass campaigns to targeted vaccination of reported at-risk populations. Funding Supported by the Bill and Melinda Gates Foundation (INV-039481).
Collapse
Affiliation(s)
- Upendo L. Mseka
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
- Public Health Institute of Malawi, Lilongwe, Malawi
| | | | | | - Oscar Divala
- Public Health Institute of Malawi, Lilongwe, Malawi
| | | | | | | | | | - Marc Y.R. Henrion
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Deus Thindwa
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
| | - Memory Mvula
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
| | - Bright Odala
- Public Health Institute of Malawi, Lilongwe, Malawi
| | | | | | - Khuzwayo C. Jere
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Kamuzu University of Health Sciences (formerly University of Malawi, College of Medicine) Blantyre, Malawi
| | - Nicholas Feasey
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Antonia Ho
- University of Glasgow, Glasgow, United Kingdom
| | - Abena S. Amoah
- London School of Hygiene and Tropical Medicine, London, United Kingdom
- Malawi Epidemiology and Intervention Unit, Lilongwe, Malawi
| | - Melita Gordon
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Todd D. Swarthout
- NIHR Mucosal Pathogens Research Unit, Research Department of Infection, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Amelia Crampin
- University of Glasgow, Glasgow, United Kingdom
- London School of Hygiene and Tropical Medicine, London, United Kingdom
- Malawi Epidemiology and Intervention Unit, Lilongwe, Malawi
| | - Robert S. Heyderman
- NIHR Mucosal Pathogens Research Unit, Research Department of Infection, Division of Infection and Immunity, University College London, London, United Kingdom
| | | | | | | | - John Phuka
- Kamuzu University of Health Sciences (formerly University of Malawi, College of Medicine) Blantyre, Malawi
| | | | | | - Kondwani C. Jambo
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Kamuzu University of Health Sciences (formerly University of Malawi, College of Medicine) Blantyre, Malawi
| | | |
Collapse
|
99
|
Prabhu M, Yang YJ, Johnston CD, Murphy EA, Ketas TJ, Diaz-Tapia R, Jurkiewicz M, Racine-Brzostek S, Mohammed I, Sukhu AC, Singh S, Forlenza K, Iyer S, Yee J, Eng D, Marks K, Zhao Z, Klasse PJ, Permar S, Moore JP, Riley LE. Longitudinal antibody response kinetics following SARS-CoV-2 messenger RNA vaccination in pregnant and nonpregnant persons. Am J Obstet Gynecol MFM 2023; 5:100796. [PMID: 36334723 PMCID: PMC9626404 DOI: 10.1016/j.ajogmf.2022.100796] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/10/2022] [Accepted: 10/28/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND For some vaccine-preventable diseases, the immunologic response to vaccination is altered by a pregnant state. The effect of pregnancy on SARS-CoV-2 vaccine response remains unclear. OBJECTIVE We sought to characterize the peak and longitudinal anti-S immunoglobulin G, immunoglobulin M, and immunoglobulin A responses to messenger RNA-based SARS-CoV-2 vaccination in pregnant persons and compare them with those in nonpregnant, reproductive-aged persons. STUDY DESIGN We conducted 2 parallel prospective cohort studies among pregnant and nonpregnant persons who received SARS-CoV-2 messenger RNA vaccinations. Blood was collected at the time of first and second vaccine doses, 2 weeks post second dosage, and with serial longitudinal follow-up up to 41.7 weeks post vaccination initiation. Anti-S immunoglobulin M, immunoglobulin G, and immunoglobulin A were analyzed by enzyme-linked immunosorbent assay. We excluded those with previous evidence of SARS-CoV-2 infection by history or presence of antinucleocapsid antibodies. In addition, for this study, we did not include individuals who received a third or booster vaccine dosage during the study period. We also excluded pregnant persons who were not fully vaccinated (14 days post receipt of the second vaccine dosage) by time of delivery and nonpregnant persons who became pregnant through the course of the study. We studied the effect of gestational age at vaccination on the anti-S response using Spearman correlation. We compared the peak anti-S antibody responses between pregnant and nonpregnant persons using a Mann-Whitney U test. We visualized and studied the longitudinal anti-S antibody response using locally weighted scatterplot smoothing, Mann-Whitney U test, and mixed analysis of variance test. RESULTS Data from 53 pregnant and 21 nonpregnant persons were included in this analysis. The median (interquartile range) age of the pregnant and nonpregnant participants was 35.0 (33.3-37.8) years and 36.0 (33.0-41.0) years, respectively. Six (11.3%) participants initiated vaccination in the first trimester, 23 (43.3%) in the second trimester, and 24 (45.3%) in the third trimester, with a median gestational age at delivery of 39.6 (39.0-40.0) weeks. The median (interquartile range) follow-up time from vaccine initiation to the last blood sample collected was 25.9 (11.9) weeks and 28.9 (12.9) weeks in the pregnant and nonpregnant cohort, respectively. Among pregnant persons, anti-S immunoglobulin G, immunoglobulin A, and immunoglobulin M responses were not associated with gestational age at vaccine initiation (all P>.05). The anti-S immunoglobulin G response at 2 weeks post second dosage was not statistically different between pregnant and nonpregnant persons (P>.05). However, the anti-S immunoglobulin M and immunoglobulin A responses at 2 weeks post second dosage were significantly higher in nonpregnant persons (P<.001 for both). The anti-S immunoglobulin G and immunoglobulin M levels 6 to 8 months after vaccine initiation fell to comparable proportions of the peak 2 weeks post second dosage antibody levels between pregnant and nonpregnant persons (immunoglobulin G P=.77; immunoglobulin M P=.51). In contrast, immunoglobulin A levels 6 to 8 months after vaccine initiation fell to statistically significantly higher proportions of peak 2 weeks post second dosage antibody levels in pregnant compared with nonpregnant persons (P=.002). Maternal anti-S immunoglobulin G levels were strongly correlated with umbilical cord anti-S immunoglobulin G levels (R=0.8, P<.001). CONCLUSION The anti-S immunoglobulin A, immunoglobulin M, and immunoglobulin G response to SARS-CoV-2 vaccination in pregnancy is independent of gestational age of vaccine initiation. Maintenance of the immunoglobulin G response is comparable between pregnant and nonpregnant persons. The differential peak response of immunoglobulin M and immunoglobulin A and the differential decline of anti-S immunoglobulin A between pregnant and nonpregnant persons requires further investigation.
Collapse
Affiliation(s)
- Malavika Prabhu
- Department of Obstetrics & Gynecology, Weill Cornell Medicine, New York, NY (Dr Prabhu, Mr Mohammed, and Dr Riley)
| | - Yawei J Yang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY (Drs Yang, Murphy, Racine-Brzostek, and Zhao); Department of Pathology and Laboratory Medicine, New York Presbyterian/Weill Cornell Medical Center, New York, NY (Drs Yang and Racine-Brzostek, Ms Sukhu, Mr Yee, Ms Eng, and Dr Zhao).
| | - Carrie D Johnston
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY (Drs Johnston and Marks)
| | - Elisabeth A Murphy
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY (Drs Yang, Murphy, Racine-Brzostek, and Zhao)
| | - Thomas J Ketas
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY (Messrs Ketas and Diaz-Tapia and Drs Klasse and Moore)
| | - Randy Diaz-Tapia
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY (Messrs Ketas and Diaz-Tapia and Drs Klasse and Moore)
| | - Magdalena Jurkiewicz
- Department of Pathology and Cell Biology, Columbia University, New York, NY (Dr Jurkiewicz)
| | - Sabrina Racine-Brzostek
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY (Drs Yang, Murphy, Racine-Brzostek, and Zhao); Department of Pathology and Laboratory Medicine, New York Presbyterian/Weill Cornell Medical Center, New York, NY (Drs Yang and Racine-Brzostek, Ms Sukhu, Mr Yee, Ms Eng, and Dr Zhao)
| | - Iman Mohammed
- Department of Obstetrics & Gynecology, Weill Cornell Medicine, New York, NY (Dr Prabhu, Mr Mohammed, and Dr Riley)
| | - Ashley C Sukhu
- Department of Pathology and Laboratory Medicine, New York Presbyterian/Weill Cornell Medical Center, New York, NY (Drs Yang and Racine-Brzostek, Ms Sukhu, Mr Yee, Ms Eng, and Dr Zhao)
| | - Sunidhi Singh
- Weill Cornell Medicine, New York, NY (Ms Singh, Dr Forlenza, and Ms Iyer)
| | - Kimberly Forlenza
- Weill Cornell Medicine, New York, NY (Ms Singh, Dr Forlenza, and Ms Iyer)
| | - Sonali Iyer
- Weill Cornell Medicine, New York, NY (Ms Singh, Dr Forlenza, and Ms Iyer)
| | - Jim Yee
- Department of Pathology and Laboratory Medicine, New York Presbyterian/Weill Cornell Medical Center, New York, NY (Drs Yang and Racine-Brzostek, Ms Sukhu, Mr Yee, Ms Eng, and Dr Zhao)
| | - Dorothy Eng
- Department of Pathology and Laboratory Medicine, New York Presbyterian/Weill Cornell Medical Center, New York, NY (Drs Yang and Racine-Brzostek, Ms Sukhu, Mr Yee, Ms Eng, and Dr Zhao)
| | - Kristen Marks
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY (Drs Johnston and Marks)
| | - Zhen Zhao
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY (Drs Yang, Murphy, Racine-Brzostek, and Zhao); Department of Pathology and Laboratory Medicine, New York Presbyterian/Weill Cornell Medical Center, New York, NY (Drs Yang and Racine-Brzostek, Ms Sukhu, Mr Yee, Ms Eng, and Dr Zhao)
| | - Per Johan Klasse
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY (Messrs Ketas and Diaz-Tapia and Drs Klasse and Moore)
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY (Dr Permar)
| | - John P Moore
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY (Messrs Ketas and Diaz-Tapia and Drs Klasse and Moore)
| | - Laura E Riley
- Department of Obstetrics & Gynecology, Weill Cornell Medicine, New York, NY (Dr Prabhu, Mr Mohammed, and Dr Riley)
| |
Collapse
|
100
|
Matkowska-Kocjan A, Owoc-Lempach J, Ludwikowska K, Szenborn F, Moskwa N, Kurek K, Kałwak K, Szenborn L, Ussowicz M. COVID-19 mRNA Vaccine Tolerance and Immunogenicity in Hematopoietic Stem Cell Transplantation Recipients Aged 5-11 Years Old-Non-Randomized Clinical Trial. Vaccines (Basel) 2023; 11:vaccines11010195. [PMID: 36680039 PMCID: PMC9866698 DOI: 10.3390/vaccines11010195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/28/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
The SARS-CoV-2 pandemic had a devastating impact on the world’s population in the years 2020−2022. The rapid development of vaccines enabled a reduction in the mortality and morbidity of COVID-19, but there are limited data about their effects on immunocompromised children. The aim of this prospective study was to evaluate the safety and efficacy of the mRNA BNT162b2 (Pfizer/Biontech) vaccine in allogeneic hematopoietic stem cell transplantation (allo-HSCT) recipients. Material and methods: Two cohorts of 34 children after allo-HSCT and 35 healthy children aged 5−11 years were vaccinated with two doses of the mRNA BNT162b2 (10 µg) vaccine. All children were evaluated for adverse effects with electronic surveys and the immunogenicity of the vaccine was assessed with anti-SARS-CoV-2 IgG titer measurements. Results: All reported adverse events (AEs) were classified as mild. The most common AE was pain at the injection site. All the other AEs (both local and systemic) were rarely reported (<15% patients). Both groups showed a similar response in anti-SARS-CoV-2 IgG production. Patients after allo-HSCT that were undergoing immunosuppressive treatment presented a poorer immunological response than patients off of treatment. Time since HSCT, patient age, lymphocyte count, and total IgG concentration did not correlate with initial/post-vaccination anti-SARS-CoV-2 IgG titers. Most patients who were eligible for a third dose of the vaccine had an excellent humoral response observed after two vaccine doses. Conclusions: The COVID-19 mRNA BNT162b2 vaccine is very well tolerated and highly immunogenic in 5−11-year-old children after HSCT. Children >2 years of age after HSCT who did not receive immunosuppressive treatment presented excellent antibody production after two doses of the vaccine, but children on immunosuppression may require a more intense vaccination schedule.
Collapse
Affiliation(s)
- Agnieszka Matkowska-Kocjan
- Department and Clinic of Pediatric Infectious Diseases, Wroclaw Medical University, 50-368 Wrocław, Poland
- Correspondence:
| | - Joanna Owoc-Lempach
- Department and Clinic of Paediatric Oncology, Haematology and Bone Marrow Transplantation, Wroclaw Medical University, 50-556 Wrocław, Poland
| | - Kamila Ludwikowska
- Department and Clinic of Pediatric Infectious Diseases, Wroclaw Medical University, 50-368 Wrocław, Poland
| | - Filip Szenborn
- Faculty of Electronics, Wroclaw University of Science and Technology, 50-370 Wrocław, Poland
| | - Natalia Moskwa
- Department and Clinic of Pediatric Infectious Diseases, Wroclaw Medical University, 50-368 Wrocław, Poland
| | - Katarzyna Kurek
- Department and Clinic of Pediatric Infectious Diseases, Wroclaw Medical University, 50-368 Wrocław, Poland
| | - Krzysztof Kałwak
- Department and Clinic of Paediatric Oncology, Haematology and Bone Marrow Transplantation, Wroclaw Medical University, 50-556 Wrocław, Poland
| | - Leszek Szenborn
- Department and Clinic of Pediatric Infectious Diseases, Wroclaw Medical University, 50-368 Wrocław, Poland
| | - Marek Ussowicz
- Department and Clinic of Paediatric Oncology, Haematology and Bone Marrow Transplantation, Wroclaw Medical University, 50-556 Wrocław, Poland
| |
Collapse
|