1
|
Lustig Y, Barda N, Weiss-Ottolenghi Y, Indenbaum V, Margalit I, Asraf K, Doolman R, Chalkias S, Das R, Elfatarany G, Harats D, Kreiss Y, Regev-Yochay G. Humoral response superiority of the monovalent XBB.1.5 over the bivalent BA.1 and BA.5 mRNA COVID-19 vaccines. Vaccine 2024; 42:126010. [PMID: 38806352 DOI: 10.1016/j.vaccine.2024.05.058] [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: 03/25/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 05/30/2024]
Abstract
JN.1, the dominating SARS-CoV-2 variant, is antigenically distinct from ancestral BA.1, BA.5 and XBB.1.5 variants, raising concern about effectiveness of updated COVID-19 vaccines. Here, we compared the neutralizing antibody response against JN.1, 1-month after receipt of the three available Moderna mRNA vaccines. Sera obtained from 37, 30 and 30 XBB.1.5, BA.1 and BA.4-5 -vaccine recipients, respectively, were tested for anti-RBD IgG and for JN-1 specific neutralizing antibody levels. Geometric mean fold rise (GMFR) in JN.1 specific neutralizing titers was 27 (95 % CI: 17-43.1), 10.1 (95 % CI: 6.48-15.7) and 8.77 (95 % CI: 5.69-13.5) following XBB.1.5, BA.1 and BA.4-5 vaccines, respectively, translating into a 64 % lower adjusted response (geometric mean ratio [GMR] = 0.36, 95 % CI: 0.21-0.6) in the BA.1 arm, and a 75 % lower response (GMR = 0.25, 95 % CI: 0.15-0.43) in the BA.4-5 arm. This suggests that XBB.1.5 vaccination will most likely, result in improved effectiveness against JN.1 compared with other COVID-19 vaccines.
Collapse
MESH Headings
- Humans
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- Female
- Male
- Immunity, Humoral
- Middle Aged
- Adult
- mRNA Vaccines/immunology
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Aged
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Immunogenicity, Vaccine
Collapse
Affiliation(s)
- Yaniv Lustig
- Central Virology Laboratory, Ministry of Health and Sheba Medical Center, Tel-Hashomer, Israel; Faculty of Medical and Health Sciences, Tel-Aviv University, Israel; The Sheba Pandemic Preparedness Research Institute (SPRI), Sheba Medical Center Tel Hashomer, Ramat Gan, Israel.
| | - Noam Barda
- The Sheba Pandemic Preparedness Research Institute (SPRI), 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 Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Yael Weiss-Ottolenghi
- The Sheba Pandemic Preparedness Research Institute (SPRI), Sheba Medical Center Tel Hashomer, Ramat Gan, Israel
| | - Victoria Indenbaum
- Central Virology Laboratory, Ministry of Health and Sheba Medical Center, Tel-Hashomer, Israel
| | - Ili Margalit
- Faculty of Medical and Health Sciences, Tel-Aviv University, Israel; The Sheba Pandemic Preparedness Research Institute (SPRI), Sheba Medical Center Tel Hashomer, Ramat Gan, Israel
| | - Keren Asraf
- Sheba Medical Center Tel Hashomer, Ramat Gan, Israel
| | - Ram Doolman
- Sheba Medical Center Tel Hashomer, Ramat Gan, Israel
| | - Spyros Chalkias
- Clinical Development, Infectious Diseases Unit, Moderna, Israel
| | - Rituparna Das
- Clinical Development, Infectious Diseases Unit, Moderna, Israel
| | | | - Dror Harats
- Sheba Medical Center Tel Hashomer, Ramat Gan, Israel
| | | | - Gili Regev-Yochay
- Faculty of Medical and Health Sciences, Tel-Aviv University, Israel; The Sheba Pandemic Preparedness Research Institute (SPRI), Sheba Medical Center Tel Hashomer, Ramat Gan, Israel
| |
Collapse
|
2
|
Durier C, Ninove L, van der Werf S, Lefebvre M, Desaint C, Bauer R, Attia M, Lecompte AS, Lachatre M, Maakaroun-Vermesse Z, Nicolas JF, Verdon R, Kiladjian JJ, Loubet P, Schmidt-Mutter C, Corbin V, Ansart S, Melica G, Resch M, Netzer E, Kherabi Y, Tardieu R, Lelièvre JD, Tartour E, Meyer L, de Lamballerie X, Launay O. Incidence of COVID-19 mRNA vaccine symptomatic breakthrough infections during Omicron circulation in adults with or without infection prior to vaccination. Infect Dis Now 2024; 54:104886. [PMID: 38494117 DOI: 10.1016/j.idnow.2024.104886] [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: 10/13/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
OBJECTIVES COVID-19 vaccine breakthrough infections were frequently reported during circulation of the Omicron variant. The ANRS|MIE CoviCompareP study investigated these infections in adults vaccinated and boosted with BNT162b2 [Pfizer-BioNTech] and with/without SARS-CoV-2 infection before vaccination. METHODS In the first half of 2021, healthy adults (aged 18-45, 65-74 and 75 or older) received either one dose of BNT162b2 (n = 120) if they had a documented history of SARS-CoV-2 infection at least five months previously, or two doses (n = 147) if they had no history confirmed by negative serological tests. A first booster dose was administered at least 6 months after the primary vaccination, and a second booster dose, if any, was reported in the database. Neutralizing antibodies (NAbs) against the European (D614G) strain and the Omicron BA.1 variant were assessed up to 28 days after the first booster dose. A case-control analysis was performed for the 252 participants who were followed up in 2022, during the Omicron waves. RESULTS From January to October 2022, 78/252 (31%) had a documented symptomatic breakthrough infection after full vaccination: 21/117 (18%) in those who had been infected before vaccination vs. 57/135 (42%) in those who had not. In a multivariate logistic regression model, factors associated with a lower risk of breakthrough infection were older age, a higher number of booster doses, and higher levels of Omicron BA.1 NAb titers in adults with infection before vaccination, but not in those without prior infection. CONCLUSION Our results highlight the need to consider immune markers of protection in association with infection and vaccination history.
Collapse
Affiliation(s)
| | - Laetitia Ninove
- Unité des Virus Émergents (UVE), Aix Marseille Univ, IRD 190, INSERM 1207, Marseille, France
| | - Sylvie van der Werf
- Institut Pasteur, Université Paris Cité, UMR 3569 CNRS, Unité de Génétique Moléculaire des Virus à ARN, Centre National de Référence Virus des Infections Respiratoires, Paris, France
| | - Maeva Lefebvre
- Service de maladies infectieuses et tropicales, Centre de prévention des maladies infectieuses et transmissibles CHU de Nantes - CIC1413 Nantes, Nantes, France
| | - Corinne Desaint
- INSERM US19, Villejuif, France; INSERM CIC 1417 Cochin Pasteur, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Innovative Clinical Research Network in Vaccinology, Université de Paris, Sorbonne Paris Cité, Paris, France
| | | | - Mikael Attia
- Institut Pasteur, Université Paris Cité, UMR 3569 CNRS, Unité de Génétique Moléculaire des Virus à ARN, Centre National de Référence Virus des Infections Respiratoires, Paris, France
| | - Anne-Sophie Lecompte
- Service de maladies infectieuses et tropicales, Centre de prévention des maladies infectieuses et transmissibles CHU de Nantes - CIC1413 Nantes, Nantes, France
| | - Marie Lachatre
- INSERM CIC 1417 Cochin Pasteur, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Innovative Clinical Research Network in Vaccinology, Université de Paris, Sorbonne Paris Cité, Paris, France
| | - Zoha Maakaroun-Vermesse
- Centre de Vaccination CHU de Tours, Centre d'Investigation Clinique CIC 1415, INSERM, CHRU de Tours, Tours, France
| | - Jean-François Nicolas
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Université Claude Bernard Lyon I, Lyon, France; CHU Lyon-Sud, Pierre-Bénite, France
| | - Renaud Verdon
- Service de Maladies Infectieuses, CHU de Caen, Dynamicure INSERM, UMR 1311, Normandie Univ, UNICAEN, Caen, France
| | - Jean-Jacques Kiladjian
- AP-HP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM, CIC1427, Université Paris Cité, Paris, France
| | - Paul Loubet
- VBMI, INSERM U1047, Department of Infectious and Tropical Diseases, Université de Montpellier, CHU Nîmes, Montpellier, France
| | | | - Violaine Corbin
- CHU Clermont-Ferrand, INSERM CIC1405, Clermont-Ferrand, France
| | | | - Giovanna Melica
- Service d'Immunologie Clinique et Maladies Infectieuses, APHP, Hôpital Henri Mondor, INSERM CIC 1430, Créteil, France
| | | | | | - Yousra Kherabi
- INSERM CIC 1417 Cochin Pasteur, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Innovative Clinical Research Network in Vaccinology, Université de Paris, Sorbonne Paris Cité, Paris, France
| | | | | | - Eric Tartour
- APHP, Hôpital Européen Georges Pompidou, INSERM U970, PARCC, Université de Paris, Paris, France
| | - Laurence Meyer
- INSERM US19, Villejuif, France; INSERM, CESP U1018, Université Paris Saclay, APHP, Le Kremlin-Bicêtre, France
| | - Xavier de Lamballerie
- Unité des Virus Émergents (UVE), Aix Marseille Univ, IRD 190, INSERM 1207, Marseille, France
| | - Odile Launay
- INSERM CIC 1417 Cochin Pasteur, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Innovative Clinical Research Network in Vaccinology, Université de Paris, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
3
|
Brummelman J, Suárez-Hernández S, de Rond L, Bogaard-van Maurik M, Molenaar P, van Wijlen E, Oomen D, Beckers L, Rots NY, van Beek J, Nicolaie MA, van Els CACM, Boer MC, Kaaijk P, Buisman AM, de Wit J. Distinct T cell responsiveness to different COVID-19 vaccines and cross-reactivity to SARS-CoV-2 variants with age and CMV status. Front Immunol 2024; 15:1392477. [PMID: 38774878 PMCID: PMC11106399 DOI: 10.3389/fimmu.2024.1392477] [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: 02/27/2024] [Accepted: 04/09/2024] [Indexed: 05/24/2024] Open
Abstract
Introduction Accumulating evidence indicates the importance of T cell immunity in vaccination-induced protection against severe COVID-19 disease, especially against SARS-CoV-2 Variants-of-Concern (VOCs) that more readily escape from recognition by neutralizing antibodies. However, there is limited knowledge on the T cell responses across different age groups and the impact of CMV status after primary and booster vaccination with different vaccine combinations. Moreover, it remains unclear whether age has an effect on the ability of T cells to cross-react against VOCs. Methods Therefore, we interrogated the Spike-specific T cell responses in healthy adults of the Dutch population across different ages, whom received different vaccine types for the primary series and/or booster vaccination, using IFNɣ ELISpot. Cells were stimulated with overlapping peptide pools of the ancestral Spike protein and different VOCs. Results Robust Spike-specific T cell responses were detected in the vast majority of participants upon the primary vaccination series, regardless of the vaccine type (i.e. BNT162b2, mRNA-1273, ChAdOx1 nCoV-19, or Ad26.COV2.S). Clearly, in the 70+ age group, responses were overall lower and showed more variation compared to younger age groups. Only in CMV-seropositive older adults (>70y) there was a significant inverse relation of age with T cell responses. Although T cell responses increased in all age groups after booster vaccination, Spike-specific T cell frequencies remained lower in the 70+ age group. Regardless of age or CMV status, primary mRNA-1273 vaccination followed by BNT162b2 booster vaccination showed limited booster effect compared to the BNT162b2/BNT162b2 or BNT162b2/mRNA-1273 primary-booster regimen. A modest reduction in cross-reactivity to the Alpha, Delta and Omicron BA.1, but not the Beta or Gamma variant, was observed after primary vaccination. Discussion Together, this study shows that age, CMV status, but also the primary-booster vaccination regimen influence the height of the vaccination-induced Spike-specific T cell response, but did not impact the VOC cross-reactivity.
Collapse
Affiliation(s)
- Jolanda Brummelman
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Sara Suárez-Hernández
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Lia de Rond
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Marjan Bogaard-van Maurik
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Petra Molenaar
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Emma van Wijlen
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Debbie Oomen
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Lisa Beckers
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Nynke Y. Rots
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Josine van Beek
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Mioara A. Nicolaie
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Cécile A. C. M. van Els
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Mardi C. Boer
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Patricia Kaaijk
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Anne-Marie Buisman
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Jelle de Wit
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
4
|
Underwood AP, Sølund C, Jacobsen K, Binderup A, Fernandez-Antunez C, Mikkelsen LS, Inekci D, Villadsen SL, Castruita JAS, Pinholt M, Fahnøe U, Ramirez S, Brix L, Weis N, Bukh J. Neutralizing antibody and CD8 + T cell responses following BA.4/5 bivalent COVID-19 booster vaccination in adults with and without prior exposure to SARS-CoV-2. Front Immunol 2024; 15:1353353. [PMID: 38571939 PMCID: PMC10987722 DOI: 10.3389/fimmu.2024.1353353] [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: 12/10/2023] [Accepted: 02/08/2024] [Indexed: 04/05/2024] Open
Abstract
As severe acute respiratory coronavirus 2 (SARS-CoV-2) variants continue to emerge, it is important to characterize immune responses against variants which can inform on protection efficacies following booster vaccination. In this study, neutralizing breadth and antigen-specific CD8+ T cell responses were analyzed in both infection-naïve and infection-experienced individuals following administration of a booster bivalent Wuhan-Hu-1+BA.4/5 Comirnaty® mRNA vaccine. Significantly higher neutralizing titers were found after this vaccination compared to the pre-third booster vaccination time point. Further, neutralizing breadth to omicron variants, including BA.1, BA.2, BA.5, BQ.1 and XBB.1, was found to be boosted following bivalent vaccination. SARS-CoV-2-specific CD8+ T cells were identified, but with no evidence that frequencies were increased following booster vaccinations. Spike protein-specific CD8+ T cells were the only responses detected after vaccination and non-spike-specific CD8+ T cells were only detected after infection. Both spike-specific and non-spike-specific CD8+ T cells were found at much lower frequencies than CD8+ T cells specific to cytomegalovirus (CMV), Epstein-Barr virus (EBV) and influenza (Flu). Taken together, these results show that the bivalent Wuhan-Hu-1+BA.4/5 Comirnaty® mRNA vaccine boosted the breadth of neutralization to newer SARS-CoV-2 variants and that vaccination is able to induce spike protein-specific CD8+ T cell responses, which are maintained longitudinally.
Collapse
Affiliation(s)
- Alexander P. Underwood
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Christina Sølund
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | | | - Alekxander Binderup
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Carlota Fernandez-Antunez
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Lotte S. Mikkelsen
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | | | - Signe Lysemose Villadsen
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Jose A. S. Castruita
- Department of Clinical Microbiology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Mette Pinholt
- Department of Clinical Microbiology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Ulrik Fahnøe
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Santseharay Ramirez
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | | | - Nina Weis
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| |
Collapse
|
5
|
Cherry N, Adisesh A, Burstyn I, Charlton C, Chen Y, Durand-Moreau Q, Labrèche F, Ruzycki S, Turnbull L, Zadunayski T, Yasui Y. Determinants of SARS-CoV-2 IgG response and decay in Canadian healthcare workers: A prospective cohort study. Vaccine 2024; 42:1168-1178. [PMID: 38278628 DOI: 10.1016/j.vaccine.2024.01.052] [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: 09/12/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
INTRODUCTION Healthcare workers (HCWs) from an interprovincial Canadian cohort gave serial blood samples to identify factors associated with anti-receptor binding domain (anti-RBD) IgG response to the SARS-CoV-2 virus. METHODS Members of the HCW cohort donated blood samples four months after their first SARS-CoV-2 immunization and again at 7, 10 and 13 months. Date and type of immunizations and dates of SARS-CoV-2 infection were collected at each of four contacts, together with information on immunologically-compromising conditions and current therapies. Blood samples were analyzed centrally for anti-RBD IgG and anti-nucleocapsid IgG (Abbott Architect, Abbott Diagnostics). Records of immunization and SARS-CoV-2 testing from public health agencies were used to assess the impact of reporting errors on estimates from the random-effects multivariable model fitted to the data. RESULTS 2752 of 4567 vaccinated cohort participants agreed to donate at least one blood sample. Modelling of anti-RBD IgG titer from 8903 samples showed an increase in IgG with each vaccine dose and with first infection. A decrease in IgG titer was found with the number of months since vaccination or infection, with the sharpest decline after the third dose. An immunization regime that included mRNA1273 (Moderna) resulted in higher anti-RBD IgG. Participants reporting multiple sclerosis, rheumatoid arthritis or taking selective immunosuppressants, tumor necrosis factor inhibitors, calcineurin inhibitors and antineoplastic agents had lower anti-RBD IgG. Supplementary analyses showed higher anti-RBD IgG in those reporting side-effects of vaccination, no relation of anti-RBD IgG to obesity and lower titers in women immunized in early or mid-pregnancy. Sensitivity analysis results suggested no important bias in the self-report data. CONCLUSION Creation of a prospective cohort was central to the credibility of results presented here. Serial serology assessments, with longitudinal analysis, provided effect estimates with enhanced accuracy and a clearer understanding of medical and other factors affecting response to vaccination.
Collapse
Affiliation(s)
- Nicola Cherry
- Division of Preventive Medicine, University of Alberta, 5-22 University Terrace, Edmonton, AB T6G 2T4, Canada.
| | - Anil Adisesh
- Division2 Division of Occupational Medicine, Department of Medicine, University of Toronto, C. David Naylor Building, 6 Queen's Park Crescent West, Toronto, ON M5S 3H2, Canada
| | - Igor Burstyn
- Department of Environmental and Occupational Health, Drexel University, Philadelphia, PA 19104, USA
| | - Carmen Charlton
- Alberta Precision Laboratories, 84440 112 St, Edmonton, AB T6G 2I2, Canada
| | - Yan Chen
- Department of Epidemiology & Cancer Control, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 735, Memphis, TN 38105, USA
| | - Quentin Durand-Moreau
- Division of Preventive Medicine, University of Alberta, 5-22 University Terrace, Edmonton, AB T6G 2T4, Canada
| | - France Labrèche
- Research Department, Institut de recherche Robert-Sauvé en santé et en sécurité du travail, 505 de Maisonneuve Blvd, West Montreal, QC H3A 3C2, Canada
| | - Shannon Ruzycki
- Department of Medicine, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB T2N 4N1, Canada
| | - LeeAnn Turnbull
- Alberta Precision Laboratories, 84440 112 St, Edmonton, AB T6G 2I2, Canada
| | - Tanis Zadunayski
- Division of Preventive Medicine, University of Alberta, 5-22 University Terrace, Edmonton, AB T6G 2T4, Canada
| | - Yutaka Yasui
- Department of Epidemiology & Cancer Control, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 735, Memphis, TN 38105, USA
| |
Collapse
|
6
|
Assantachai P, Niyomnaitham S, Toh ZQ, Thammasalee M, Pengsorn N, Monklang W, Licciardi PV, Chokephaibulkit K. Immunogenicity and reactogenicity of repeated intradermal mRNA COVID-19 vaccines administered as a second booster dose in a Thai geriatric population. Front Immunol 2024; 14:1302041. [PMID: 38274835 PMCID: PMC10810025 DOI: 10.3389/fimmu.2023.1302041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/21/2023] [Indexed: 01/27/2024] Open
Abstract
Background Geriatric populations are at an increased risk of severe presentations, hospitalization, and loss of life from COVID-19. Few studies have explored vaccination regimens in adults >65 years old. Repeated booster vaccination is required for high-risk populations as COVID-19 vaccine efficacy is short-lived. We compared the immunogenicity and reactogenicity of second intradermal (ID) COVID-19 booster vaccination with second intramuscular (IM) vaccination in older adults. Methods This single-center, open-labeled, prospective, cohort study conducted at Siriraj Hospital enrolled older adults ≥65 years old who previously received a first booster (third dose) mRNA vaccine (mRNA-1273 or BNT162b2) via ID or IM administration. Participants were allocated to receive a second booster of the same vaccine type and route as their first booster 16-17 weeks thereafter. Anti-SARS-CoV-2 receptor binding domain IgG and neutralizing antibody titers against Wuhan and Omicron subvariants (BA.1, BA.2, and BA.4/5) were measured 2 weeks after vaccination. Results Of 91 enrolled participants, 72.5% were women, with a median age of 75 years. Forty-nine participants (53.8%) received a second ID booster, and 42 (46.2%) received a second IM booster. Two weeks after the second booster, all groups generated anamnestic IgG antibody responses that were 5.41- to 10.00-fold higher than at baseline. Overall, higher antibody GMTs against Wuhan and Omicron subvariants were observed in IM compared with ID regimens. ID mRNA-1273 induced similar GMTs to IM BNT162b2 2 weeks after the second booster against Wuhan (486.77 [321.48, 737.05] vs. 472.63 [291.24, 767.01], respectively; p = 0.072). Higher GMTs against Omicron BA.1 (GMR [95% CI], 1.71 [1.39, 2.11]; p = 0.023), BA.2 (1.34 [1.11, 1.62]; p = 0.845), and BA.4/5 (1.10 [0.92, 1.33]; p = 0.531) were seen in all groups at 2 weeks after the second booster compared with 2-4 weeks after the first booster. Both local and systemic AEs were less frequent after the second than after the first booster, regardless of administrative route and vaccine type. Local AEs were significantly more frequent in ID mRNA-1273 arms than their respective BNT162b2 arms 2 weeks after the second booster (ID-mRNA-1273 vs. ID-BNT162b2: p ≤ 0.001). Conclusion Repeated fractional ID vaccination may be an alternative booster vaccination strategy for geriatric populations.
Collapse
Affiliation(s)
- Prasert Assantachai
- Department of Preventive and Social Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Suvimol Niyomnaitham
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Institute of Clinical Research (SICRES), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Zheng Quan Toh
- Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department of Pediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Monthira Thammasalee
- Department of Preventive and Social Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Napaporn Pengsorn
- Department of Preventive and Social Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wiyachatr Monklang
- Department of Preventive and Social Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Paul V. Licciardi
- Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department of Pediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Kulkanya Chokephaibulkit
- Siriraj Institute of Clinical Research (SICRES), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
7
|
Akhtar M, Islam MR, Khaton F, Soltana UH, Jafrin SA, Rahman SIA, Tauheed I, Ahmed T, Khan II, Akter A, Khan ZH, Islam MT, Khanam F, Biswas PK, Ahmmed F, Ahmed S, Rashid MM, Hossain MZ, Alam AN, Alamgir ASM, Rahman M, Ryan ET, Harris JB, LaRocque RC, Flora MS, Chowdhury F, Khan AI, Banu S, Shirin T, Bhuiyan TR, Qadri F. Appearance of tolerance-induction and non-inflammatory SARS-CoV-2 spike-specific IgG4 antibodies after COVID-19 booster vaccinations. Front Immunol 2023; 14:1309997. [PMID: 38173725 PMCID: PMC10763240 DOI: 10.3389/fimmu.2023.1309997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
Background Understanding the characteristics of the humoral immune responses following COVID-19 vaccinations is crucial for refining vaccination strategies and predicting immune responses to emerging SARS-CoV-2 variants. Methods A longitudinal analysis of SARS-CoV-2 spike receptor binding domain (RBD) specific IgG antibody responses, encompassing IgG subclasses IgG1, IgG2, IgG3, and IgG4 was performed. Participants received four mRNA vaccine doses (group 1; n=10) or two ChAdOx1 nCoV-19 and two mRNA booster doses (group 2; n=19) in Bangladesh over two years. Results Findings demonstrate robust IgG responses after primary Covishield or mRNA doses; declining to baseline within six months. First mRNA booster restored and surpassed primary IgG responses but waned after six months. Surprisingly, a second mRNA booster did not increase IgG levels further. Comprehensive IgG subclass analysis showed primary Covishield/mRNA vaccination generated predominantly IgG1 responses with limited IgG2/IgG3, Remarkably, IgG4 responses exhibited a distinct pattern. IgG4 remained undetectable initially but increased extensively six months after the second mRNA dose, eventually replacing IgG1 after the 3rd/4th mRNA doses. Conversely, initial Covishield recipients lack IgG4, surged post-second mRNA booster. Notably, mRNA-vaccinated individuals displayed earlier, robust IgG4 levels post first mRNA booster versus Covishield counterparts. IgG1 to IgG4 ratios decreased with increasing doses, most pronounced with four mRNA doses. This study highlights IgG response kinetics, influenced by vaccine type and doses, impacting immunological tolerance and IgG4 induction, shaping future vaccination strategies. Conclusions This study highlights the dynamics of IgG responses dependent on vaccine type and number of doses, leading to immunological tolerance and IgG4 induction, and shaping future vaccination strategies.
Collapse
Affiliation(s)
- Marjahan Akhtar
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Rashedul Islam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Fatema Khaton
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Umma Hany Soltana
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Syeda Anoushka Jafrin
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Sadia Isfat Ara Rahman
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Imam Tauheed
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Tasnuva Ahmed
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Ishtiakul Islam Khan
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Afroza Akter
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Zahid Hasan Khan
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Taufiqul Islam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Farhana Khanam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Prasanta Kumar Biswas
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Faisal Ahmmed
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Shakeel Ahmed
- Bangladesh Institute of Tropical & Infectious Diseases, Chittagong, Bangladesh
| | - Md. Mamunur Rashid
- Bangladesh Institute of Tropical & Infectious Diseases, Chittagong, Bangladesh
| | - Md. Zakir Hossain
- Bangladesh Institute of Tropical & Infectious Diseases, Chittagong, Bangladesh
| | - Ahmed Nawsher Alam
- Institute of Epidemiology, Disease Control and Research (IEDCR), Dhaka, Bangladesh
| | - A. S. M. Alamgir
- Institute of Epidemiology, Disease Control and Research (IEDCR), Dhaka, Bangladesh
| | - Mahbubur Rahman
- Institute of Epidemiology, Disease Control and Research (IEDCR), Dhaka, Bangladesh
| | - Edward T. Ryan
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Jason B. Harris
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Regina C. LaRocque
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | | | - Fahima Chowdhury
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Ashraful Islam Khan
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Sayera Banu
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Tahmina Shirin
- Institute of Epidemiology, Disease Control and Research (IEDCR), Dhaka, Bangladesh
| | - Taufiqur Rahman Bhuiyan
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Firdausi Qadri
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| |
Collapse
|
8
|
Jacobsen H, Sitaras I, Katzmarzyk M, Cobos Jiménez V, Naughton R, Higdon MM, Deloria Knoll M. Systematic review and meta-analysis of the factors affecting waning of post-vaccination neutralizing antibody responses against SARS-CoV-2. NPJ Vaccines 2023; 8:159. [PMID: 37863890 PMCID: PMC10589259 DOI: 10.1038/s41541-023-00756-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/02/2023] [Indexed: 10/22/2023] Open
Abstract
Mass COVID-19 vaccination and continued introduction of new SARS-CoV-2 variants increased prevalence of hybrid immunity at various stages of waning protection. We systematically reviewed waning of post-vaccination neutralizing antibody titers in different immunological settings to investigate differences. We searched published and pre-print studies providing post-vaccination neutralizing antibody responses against the Index strain or Omicron BA.1. We used random effects meta-regression to estimate fold-reduction from months 1 to 6 post last dose by primary vs booster regimen and infection-naïve vs hybrid-immune cohorts. Among 26 eligible studies, 65 cohorts (range 3-21 per stratum) were identified. Month-1 titers varied widely across studies within each cohort and by vaccine platform, number of doses and number of prior infections. In infection-naïve cohorts, the Index strain waned 5.1-fold (95%CI: 3.4-7.8; n = 19 cohorts) post-primary regimen and 3.8-fold (95%CI: 2.4-5.9; n = 21) post-booster from months 1 to 6, and against Omicron BA.1 waned 5.9-fold (95%CI: 3.8-9.0; n = 16) post-booster; Omicron BA.1 titers post-primary were too low to assess. In hybrid-immune, post-primary cohorts, titers waned 3.7-fold (95%CI: 1.7-7.9; n = 8) against the Index strain and 5.0-fold (95%CI: 1.1-21.8; n = 6) against Omicron BA.1; post-booster studies of hybrid-immune cohorts were too few (n = 3 cohorts each strain) to assess. Waning was similar across vaccination regimen and prior-infection status strata but was faster for Omicron BA.1 than Index strains, therefore, more recent sub-variants should be monitored. Wide differences in peak titers by vaccine platform and prior infection status mean titers drop to non-protective levels sooner in some instances, which may affect policy.
Collapse
Affiliation(s)
- Henning Jacobsen
- Department of Viral Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany.
| | - Ioannis Sitaras
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | | | | | - Melissa M Higdon
- International Vaccine Access Center, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Maria Deloria Knoll
- International Vaccine Access Center, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
9
|
Edelstein M, Wiegler Beiruti K, Ben-Amram H, Beer N, Sussan C, Batya P, Zarka S, Abu Jabal K. Vaccine-induced and hybrid immunity to SARS-CoV-2 after three or four doses of BNT162b2 - results from 22 months follow-up of a healthcare workers cohort, Israel, 2020-2022. Int J Infect Dis 2023; 135:57-62. [PMID: 37572957 DOI: 10.1016/j.ijid.2023.08.009] [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: 05/30/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023] Open
Abstract
OBJECTIVES SARS-CoV-2 remains a global health concern 3 years after its emergence. Safe and effective vaccines mitigate the pandemic impact, but the optimal schedule remains unclear, especially in a context where a high proportion of the population is infected. METHODS We periodically measured anti-spike SARS-CoV-2 immunoglobulin (Ig)G titers using a quantitative assay in an Israeli healthcare worker cohort who all received at least two BNT162b2 doses and either received further doses and/or were subsequently infected up to 22 months after dose two, and compared geometric mean concentrations according to number of doses received and infection status using analysis of variance. RESULTS Among the 993 included participants, infection after dose two led to higher geometric mean concentration IgG titers than a third dose (4285 vs 2845 arbitrary unit/ml 1-2 months after infection/vaccination, P = 0.03). In 16-18 months after dose two, those infected and those who received three or four vaccine doses all had IgG geometric mean concentration levels above 500 arbitrary unit/ml with no significant differences among groups (P = 0.6). IgG levels plateaued 16-22 months after dose two. CONCLUSION Three BNT162b2 doses provide long-term immunogenicity comparable to breakthrough infection after dose two. Dose four transiently increases IgG levels and may be especially important for providing additional protection to vulnerable individuals during periods of increased transmission risk.
Collapse
Affiliation(s)
- Michael Edelstein
- Ziv Medical Center, Safed, Israel; Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
| | | | | | | | | | | | - Salman Zarka
- Ziv Medical Center, Safed, Israel; Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Kamal Abu Jabal
- Ziv Medical Center, Safed, Israel; Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
10
|
Gonen T, Barda N, Asraf K, Joseph G, Weiss-Ottolenghi Y, Doolman R, Kreiss Y, Lustig Y, Regev-Yochay G. Immunogenicity and Reactogenicity of Coadministration of COVID-19 and Influenza Vaccines. JAMA Netw Open 2023; 6:e2332813. [PMID: 37682571 PMCID: PMC10492184 DOI: 10.1001/jamanetworkopen.2023.32813] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/01/2023] [Indexed: 09/09/2023] Open
Abstract
Importance COVID-19 and seasonal influenza vaccines were previously given separately, although their coadministration is warranted for vaccination adherence. Limited data on their coadministration have been published. Objective To compare the reactogenicity and immunogenicity of COVID-19 and influenza vaccinations administered together with those of COVID-19 vaccination alone. Design, Setting, and Participants This prospective cohort study included health care workers at a large tertiary medical center in Israel who received the Influvac Tetra (Abbott) influenza vaccine (2022/2023), the Omicron BA.4/BA.5-adapted bivalent (Pfizer/BioNTech) vaccine, or both. Vaccination began in September 2022, and data were collected until January 2023. Vaccines were offered to all employees and were coadministered or given separately. Adverse reaction questionnaires were sent, and serologic samples were also collected. Exposures Receiving COVID-19 vaccine, influenza vaccine, or both. Main Outcomes and Measures The main outcomes for the reactogenicity analysis were symptoms following vaccine receipt, assessed by a digital questionnaire: any local symptoms; fever; weakness or fatigue; any systemic symptoms; and their duration. The immunogenicity analysis' outcome was postvaccination anti-spike IgG titer. Results This study included 2 cohorts for 2 separate analyses. The reactogenicity analysis included 588 participants (of 649 questionnaire responders): 85 in the COVID-19 vaccine-alone group (median [IQR] age, 71 [58-74] years; 56 [66%] female); 357 in the influenza vaccine-alone group (median [IQR] age, 55 [40-65] years; 282 [79%] female); and 146 in the coadministration group (median [IQR] age, 61 [50-71] years; 81 [55%] female). The immunogenicity analysis included 151 participants: 74 participants in the COVID-19 vaccine group (median [IQR] age, 67 [56-73] years; 45 [61%] female) and 77 participants in the coadministration group (median [IQR] age, 60 [49-73] years; 42 [55%] female). Compared with COVID-19 vaccination alone, the risk of systemic symptoms was similar in the coadministration group (odds ratio, 0.82; 95% CI, 0.43-1.56). Geometric mean titers in the coadministration group were estimated to be 0.84 (95% CI, 0.69-1.04) times lower than in the COVID-19 vaccine-alone group. Conclusions and Relevance In this cohort study of health care workers who received a COVID-19 vaccine, an influenza vaccine, or both, coadministration was not associated with substantially inferior immune response or to more frequent adverse events compared with COVID-19 vaccine administration alone, supporting the coadministration of these vaccines.
Collapse
Affiliation(s)
- Tal Gonen
- Sheba Pandemic Research Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- The Infection Prevention & Control Unit, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Noam Barda
- Sheba Pandemic Research Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- 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
| | - Keren Asraf
- The Dworman Automated-Mega Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Gili Joseph
- Sheba Pandemic Research Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- The Infection Prevention & Control Unit, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Yael Weiss-Ottolenghi
- Sheba Pandemic Research Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- The Infection Prevention & Control Unit, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ram Doolman
- The Dworman Automated-Mega Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Yitshak Kreiss
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
- General Management, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Yaniv Lustig
- Sheba Pandemic Research Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- 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
- Sheba Pandemic Research Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- The Infection Prevention & Control Unit, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
11
|
Rosa RG, Falavigna M, Manfio JL, de Araujo CLP, Cohen M, do Valle Barbosa GRG, de Souza AP, Romeiro Silva FK, Sganzerla D, da Silva MMD, Ferreira D, de Oliveira Rodrigues C, de Souza EM, de Oliveira JC, Gradia DF, Brandalize APC, Royer CA, Luiz RM, Kucharski GA, Pedrotti F, Valluri SR, Srivastava A, Julião VW, Melone OC, Allen KE, Kyaw MH, Spinardi J, Del Carmen Morales Castillo G, McLaughlin JM. BNT162b2 mRNA COVID-19 against symptomatic Omicron infection following a mass vaccination campaign in southern Brazil: A prospective test-negative design study. Vaccine 2023; 41:5461-5468. [PMID: 37507274 DOI: 10.1016/j.vaccine.2023.07.038] [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: 04/23/2023] [Revised: 07/01/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Evidence regarding effectiveness of BNT162b2 mRNA COVID-19 vaccine against Omicron in Latin America is limited. We estimated BNT162b2 effectiveness against symptomatic COVID-19 in Brazil when Omicron was predominant. METHODS This prospective test-negative, case-control study was conducted in Toledo, Brazil, following a mass COVID-19 vaccination with BNT162b2. Patients were included if they were aged ≥12 years, sought care for acute respiratory symptoms in the public health system between November 3, 2021 and June 20, 2022, and were tested for SARS-CoV-2 using RT-PCR. In the primary analysis, we determined the effectiveness of two doses of BNT162b2 against symptomatic COVID-19. RESULTS A total of 4,574 were enrolled; of these, 1,758 patients (586 cases and 1,172 controls) were included in the primary analysis. Mean age was 27.7 years, 53.8 % were women, and 90.1 % had a Charlson comorbidity index of zero. Omicron accounted for >97 % of all identified SARS-CoV-2 variants, with BA.1 and BA.2 accounting for 84.3 % and 12.6 %, respectively. Overall adjusted estimate of two-dose vaccine effectiveness against symptomatic COVID-19 was 46.7 % (95 %CI, 19.9 %-64.6 %) after a median time between the second dose and the beginning of COVID-19 symptoms of 94 days (IQR, 60-139 days). Effectiveness waned from 77.7 % at 7-29 days after receipt of a second dose to <30 % (non-significant) after ≥120 days. CONCLUSION In a relatively young and healthy Brazilian population, two doses of BNT162b2 provided protection against symptomatic Omicron infection. However, this protection waned significantly over time, underscoring the need for boosting with variant-adapted vaccines in this population prior to waves of disease activity. TRIAL REGISTRATION NUMBER ClinicalTrials.gov number, NCT05052307 (https://clinicaltrials.gov/ct2/show/NCT05052307).
Collapse
Affiliation(s)
- Regis Goulart Rosa
- Internal Medicine Department, Hospital Moinhos de Vento (HMV), Porto Alegre, RS, Brazil; Research Unit, Inova Medical, Porto Alegre, RS, Brazil; Research Institute, HMV, Porto Alegre, RS, Brazil.
| | - Maicon Falavigna
- Research Unit, Inova Medical, Porto Alegre, RS, Brazil; Research Institute, HMV, Porto Alegre, RS, Brazil; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | | | | | - Mírian Cohen
- Research Institute, HMV, Porto Alegre, RS, Brazil; Federal University of Rio Grande do Sul (UFRGS), Brazil
| | | | | | | | | | | | | | | | | | | | - Daniela Fiori Gradia
- Department of Biochemistry and Molecular Biology, Department of Genetics - UFPR, Brazil
| | | | - Carla Adriane Royer
- Department of Biochemistry and Molecular Biology, Department of Genetics - UFPR, Brazil
| | - Rafael Messias Luiz
- Faculty of Medicine - Campus Toledo - Federal University of Paraná (UFPR), Brazil
| | | | | | - Srinivas Rao Valluri
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| | - Amit Srivastava
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA; Orbital Therapeutics, Cambridge, MA, USA
| | - Viviane Wal Julião
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| | - Olga Chameh Melone
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| | - Kristen E Allen
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| | - Moe H Kyaw
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| | - Julia Spinardi
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| | | | - John M McLaughlin
- Pfizer, Vaccines Medical and Scientific Affairs - Emerging Markets, Collegeville, PA, USA
| |
Collapse
|
12
|
Zhou W, Jiang L, Liao S, Wu F, Yang G, Hou L, Liu L, Pan X, Jia W, Zhang Y. Vaccines' New Era-RNA Vaccine. Viruses 2023; 15:1760. [PMID: 37632102 PMCID: PMC10458896 DOI: 10.3390/v15081760] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
RNA vaccines, including conventional messenger RNA (mRNA) vaccines, circular RNA (circRNA) vaccines, and self-amplifying RNA (saRNA) vaccines, have ushered in a promising future and revolutionized vaccine development. The success of mRNA vaccines in combating the COVID-19 pandemic caused by the SARS-CoV-2 virus that emerged in 2019 has highlighted the potential of RNA vaccines. These vaccines possess several advantages, such as high efficacy, adaptability, simplicity in antigen design, and the ability to induce both humoral and cellular immunity. They also offer rapid and cost-effective manufacturing, flexibility to target emerging or mutant pathogens and a potential approach for clearing immunotolerant microbes by targeting bacterial or parasitic survival mechanisms. The self-adjuvant effect of mRNA-lipid nanoparticle (LNP) formulations or circular RNA further enhances the potential of RNA vaccines. However, some challenges need to be addressed. These include the technology's immaturity, high research expenses, limited duration of antibody response, mRNA instability, low efficiency of circRNA cyclization, and the production of double-stranded RNA as a side product. These factors hinder the widespread adoption and utilization of RNA vaccines, particularly in developing countries. This review provides a comprehensive overview of mRNA, circRNA, and saRNA vaccines for infectious diseases while also discussing their development, current applications, and challenges.
Collapse
Affiliation(s)
- Wenshuo Zhou
- CNBG-Virogin Biotech (Shanghai) Co., Ltd., Shanghai 201800, China; (W.Z.); (L.J.); (S.L.); (F.W.); (G.Y.); (L.H.); (L.L.); (X.P.); (W.J.)
| | - Linglei Jiang
- CNBG-Virogin Biotech (Shanghai) Co., Ltd., Shanghai 201800, China; (W.Z.); (L.J.); (S.L.); (F.W.); (G.Y.); (L.H.); (L.L.); (X.P.); (W.J.)
| | - Shimiao Liao
- CNBG-Virogin Biotech (Shanghai) Co., Ltd., Shanghai 201800, China; (W.Z.); (L.J.); (S.L.); (F.W.); (G.Y.); (L.H.); (L.L.); (X.P.); (W.J.)
| | - Feifei Wu
- CNBG-Virogin Biotech (Shanghai) Co., Ltd., Shanghai 201800, China; (W.Z.); (L.J.); (S.L.); (F.W.); (G.Y.); (L.H.); (L.L.); (X.P.); (W.J.)
| | - Guohuan Yang
- CNBG-Virogin Biotech (Shanghai) Co., Ltd., Shanghai 201800, China; (W.Z.); (L.J.); (S.L.); (F.W.); (G.Y.); (L.H.); (L.L.); (X.P.); (W.J.)
| | - Li Hou
- CNBG-Virogin Biotech (Shanghai) Co., Ltd., Shanghai 201800, China; (W.Z.); (L.J.); (S.L.); (F.W.); (G.Y.); (L.H.); (L.L.); (X.P.); (W.J.)
| | - Lan Liu
- CNBG-Virogin Biotech (Shanghai) Co., Ltd., Shanghai 201800, China; (W.Z.); (L.J.); (S.L.); (F.W.); (G.Y.); (L.H.); (L.L.); (X.P.); (W.J.)
| | - Xinping Pan
- CNBG-Virogin Biotech (Shanghai) Co., Ltd., Shanghai 201800, China; (W.Z.); (L.J.); (S.L.); (F.W.); (G.Y.); (L.H.); (L.L.); (X.P.); (W.J.)
| | - William Jia
- CNBG-Virogin Biotech (Shanghai) Co., Ltd., Shanghai 201800, China; (W.Z.); (L.J.); (S.L.); (F.W.); (G.Y.); (L.H.); (L.L.); (X.P.); (W.J.)
- Shanghai-Virogin Biotech Co., Ltd., Shanghai 201800, China
| | - Yuntao Zhang
- CNBG-Virogin Biotech (Shanghai) Co., Ltd., Shanghai 201800, China; (W.Z.); (L.J.); (S.L.); (F.W.); (G.Y.); (L.H.); (L.L.); (X.P.); (W.J.)
- Sinopharm Group China National Biotech Group (CNBG) Co., Ltd., Beijing 100124, China
| |
Collapse
|
13
|
Beccia F, Lontano A, Rossi MF, Marziali E, Pascucci D, Raponi M, Santoro PE, Moscato U, Laurenti P. Three-year COVID-19 and flu vaccinations among medical residents in a tertiary hospital in Italy: The threat of acceptance decline in seasonal campaigns. Hum Vaccin Immunother 2023; 19:2252708. [PMID: 37706326 PMCID: PMC10503443 DOI: 10.1080/21645515.2023.2252708] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/04/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023] Open
Abstract
The COVID-19 vaccination campaign in Italy started in December 2020, and, due to the Omicron variant's emergence, a second booster dose was recommended for high-risk individuals and healthcare workers from July 2022. The aim of the study was to evaluate the vaccination coverages for the COVID-19 second booster dose and to identify predictors of its acceptance within the population of medical residents (MRs) of the Fondazione Policlinico Universitario "A. Gemelli" IRCCS (FPG) University Hospital. The study was conducted at FPG from October 4th to December 21st, 2022, and COVID-19 second booster dose and influenza vaccines were administered. The study analyzed collected data and conducted multivariate logistic regressions to explore potential predictors of vaccination adherence. The analyses performed were compared with the sample enrolled in FPG residency programs at the start of the COVID-19 vaccination campaign. 1968 MRs were involved in the 2022-2023 vaccination campaign (mean age 28.97, SD 3.44), and the second booster dose of COVID-19 vaccination coverage was low (18.80%). Almost all participants opted for co-administration of COVID-19 and influenza vaccinations, leading to a similar rate of influenza vaccination coverage (16.26%). Being a frontline resident, meaning a direct involvement in managing COVID-19 patients and vaccination campaigns, was the main predictor of vaccination adherence (OR 1.72, 95% CI 1.25-2.17). The dropping in influenza vaccination coverage in 2022-2023 and the low adherence to COVID-19 second booster dose among young physicians is concerning, calling for tailored vaccination campaigns and interventions.
Collapse
Affiliation(s)
- Flavia Beccia
- Section of Hygiene - Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alberto Lontano
- Section of Hygiene - Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Francesca Rossi
- Section of Occupational Health - Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eleonora Marziali
- Section of Hygiene - Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Domenico Pascucci
- Section of Hygiene - Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Health Management, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Matteo Raponi
- Department of Women, Child and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Paolo Emilio Santoro
- Section of Occupational Health - Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Women, Child and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Umberto Moscato
- Section of Occupational Health - Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Women, Child and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Patrizia Laurenti
- Section of Hygiene - Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Women, Child and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
14
|
Silva MFS, de Oliveira FDCE, Pinto ACMD, Luciano MCDS, Garcia MML, Araújo FMDC, Fonseca MHG. Antibody response following the fourth SARS-CoV-2 vaccine dose during the Omicron wave in Brazil. J Med Virol 2023; 95:e29055. [PMID: 37641396 DOI: 10.1002/jmv.29055] [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: 04/04/2023] [Revised: 08/03/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023]
Abstract
We investigated the impact of the fourth dose with ChAdOx1 nCoV-19 (AstraZeneca) in the humoral immune response to SARS-CoV-2 during a 9-month follow-up period in which Omicron was the predominant variant in Brazil. IgG for the SARS-CoV-2 spike protein (S) and nucleocapsid (N) proteins were analyzed in samples collected before and after the fourth dose. All participants were tested monthly for SARS-CoV-2 infection by RT-qPCR. The antibody response induced by the fourth dose of the coronavirus disease 2019 vaccine was evaluated and compared with the response induced by the second and third doses. The additional antibody response to the viral S protein after the fourth dose was smaller than those after the third vaccine dose. In contrast, an increase in the N IgG levels could be observed after the fourth dose compared to other vaccine doses. In the comparison of the antibody response before and after the fourth dose, an increase in both S-and-N IgG was noted, mainly in the positive qPCR group. We did not observe a significant decline in IgG levels after the fourth dose, as observed after the second and third doses, therefore, a sustained humoral response to both S and N proteins seems to be achieved.
Collapse
|
15
|
Gómez de la Torre JC, Hueda-Zavaleta M, Cáceres-DelAguila JA, Muro-Rojo C, Cruz-Escurra NDL, Benítes-Zapata VA. Humoral Response after a Fourth Dose with mRNA-1273 in Healthcare Workers with and without a History of SARS-CoV-2 Infection and Previously Vaccinated with Two Doses of BBIBP-CorV Plus BNT162b2 Vaccine. Vaccines (Basel) 2023; 11:vaccines11050894. [PMID: 37242998 DOI: 10.3390/vaccines11050894] [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: 01/24/2023] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 05/28/2023] Open
Abstract
There is limited information on the kinetics of the humoral response elicited by a fourth dose with a heterologous mRNA1273 booster in patients who previously received a third dose with BNT162b2 and two doses of BBIBP-CorV as the primary regimen. We conducted a prospective cohort study to assess the humoral response using Elecsys® anti-SARS-CoV-2 S (anti-S-RBD) of 452 healthcare workers (HCWs) in a private laboratory in Lima, Peru at 21, 120, 210, and 300 days after a third dose with a BNT162b2 heterologous booster in HCW previously immunized with two doses of BBIBP-CorV, depending on whether or not they received a fourth dose with the mRNA1273 heterologous vaccine and on the history of previous SARS infection -CoV-2. Of the 452 HCWs, 204 (45.13%) were previously infected (PI) with SARS-CoV-2, and 215 (47.57%) received a fourth dose with a heterologous mRNA-1273 booster. A total of 100% of HCWs presented positive anti-S-RBD 300 days after the third dose. In HCWs receiving a fourth dose, GMTs 2.3 and 1.6 times higher than controls were observed 30 and 120 days after the fourth dose. No statistically significant differences in anti-S-RBD titers were observed in those HCWs PI and NPI during the follow-up period. We observed that HCWs who received a fourth dose with the mRNA1273 and those previously infected after the third dose with BNT162b2 (during the Omicron wave) presented higher anti-S-RBD titers (5734 and 3428 U/mL, respectively). Further studies are required to determine whether patients infected after the third dose need a fourth dose.
Collapse
Affiliation(s)
| | - Miguel Hueda-Zavaleta
- Hospital III Daniel Alcides Carrión, Essalud, Calana Road, Km 6.5, Calana, Tacna 23000, Peru
- Facultad de Ciencias de la Salud, Universidad Privada de Tacna, Bolognesi Avenue Number 1177, Tacna 23003, Peru
| | | | - Cecilia Muro-Rojo
- Roe Clinical Laboratory, Dos de Mayo Avenue, 1741, San Isidro, Lima 15076, Peru
| | | | - Vicente A Benítes-Zapata
- Unidad de Investigación para la Generación y Síntesis de Evidencias en Salud, Universidad San Ignacio de Loyola, La Fontana Avenue 550, La Molina, Lima 15024, Peru
| |
Collapse
|
16
|
SARS-CoV-2 live virus neutralization after four COVID-19 vaccine doses in people with HIV receiving suppressive antiretroviral therapy. AIDS 2023; 37:F11-F18. [PMID: 36789806 PMCID: PMC9994812 DOI: 10.1097/qad.0000000000003519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
OBJECTIVE Limited data exist regarding the immune benefits of fourth COVID-19 vaccine doses in people with HIV (PWH) receiving antiretroviral therapy (ART), particularly now that most have experienced a SARS-CoV-2 infection. We quantified wild-type, Omicron-BA.5 and Omicron-BQ.1-specific neutralization up to 1 month post-fourth COVID-19 vaccine dose in 63 (19 SARS-CoV-2-naive and 44 SARS-CoV-2-experienced) PWH. DESIGN A longitudinal observational cohort. METHODS Quantification of wild-type-, Omicron-BA.5, and Omicron-BQ.1-specific neutralization using live virus assays. RESULTS Participants received monovalent (44%) and bivalent (56%) mRNA fourth doses. In COVID-19-naive PWH, fourth doses enhanced wild-type and Omicron-BA.5-specific neutralization modestly above three-dose levels ( P = 0.1). In COVID-19-experienced PWH, fourth doses enhanced wild-type specific neutralization modestly ( P = 0.1) and BA.5-specific neutralization substantially ( P = 0.002). Consistent with humoral benefits of 'hybrid' immunity, COVID-19-experienced PWH exhibited the highest neutralization post-fourth dose, wherein those with Omicron-era infections displayed higher wild-type specific ( P = 0.04) but similar BA.5 and BQ.1-specific neutralization than those with pre-Omicron-era infections. Nevertheless, BA.5-specific neutralization was significantly below wild-type in everyone regardless of COVID-19 experience, with BQ.1-specific neutralization lower still (both P < 0.0001). In multivariable analyses, fourth dose valency did not affect neutralization magnitude. Rather, an mRNA-1273 fourth dose (versus a BNT162b2 one) was the strongest correlate of wild-type specific neutralization, while prior COVID-19, regardless of pandemic era, was the strongest correlate of BA.5 and BQ.1-specific neutralization post-fourth dose. CONCLUSION Fourth COVID-19 vaccine doses, irrespective of valency, benefit PWH regardless of prior SARS-CoV-2 infection. Results support recommendations that all adults receive a fourth COVID-19 vaccine dose within 6 months of their third dose (or their most recent SARS-CoV-2 infection).
Collapse
|