1
|
Meeraus W, Postema A, Gray CM, Lee A, Maria AS, Furtado BE, Conde-Sousa E, Ouwens M, Valverde DA, da Cunha CA, Barbosa AN, Corte C, Taylor S. Second booster doses of adenoviral- and mRNA-based COVID-19 vaccines increase protection against COVID-19 hospitalization: Final analysis from the REFORCO-Brazil real-world effectiveness study during Omicron. Vaccine 2025; 53:126955. [PMID: 40068393 DOI: 10.1016/j.vaccine.2025.126955] [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: 10/21/2024] [Revised: 02/14/2025] [Accepted: 02/22/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Booster doses of COVID-19 vaccines are required to maintain protection against SARS-CoV-2. However, real-world evidence from South America, needed to inform optimal vaccination strategies, is lacking. Herein, we present the final analysis of REFORCO-Brazil, a large-scale assessment of relative vaccine effectiveness (rVE) of second boosters (vs first boosters) against hospitalization with COVID-19. METHODS REFORCO-Brazil is a test-negative case-control study (NCT05697705) that utilized Brazilian national data on severe acute respiratory syndrome (SARS) surveillance and COVID-19 vaccination. Individuals hospitalized with SARS from January 1 to December 31, 2022, were classified as test-positive cases (via SARS-CoV-2 antigen/reverse transcription polymerase chain reaction [RT-PCR]) or test-negative case-controls (via RT-PCR) and matched by admission date, region, sex, preceding COVID-19 vaccinations, and age. We used conditional logistic regression combined with multiple covariate adjustments to estimate rVE for second boosters (versus first boosters received ≥4 months prior) overall, by type (AZD1222, Ad26.COV2·S, BNT162b2, and CoronaVac) and in vulnerable subgroups (elderly and immunocompromised/high-risk individuals). RESULTS Median (range) time between second booster and SARS hospitalization was 87.0 (8.0-307.0) and 79.0 (8.0-303.0) days among 5426 test-positive cases and 6131 test-negative controls, respectively. Overall rVE of any second booster against hospitalization was 18.7 % (95 % confidence interval [CI]: 10.5-26.1). The rVE of adenoviral- and mRNA-based vaccines was similar; 18.2 % (4.8-29.8) for AZD1222, 20.7 % (10.2-30.0) for Ad26.COV2·S, and 23.2 % (9.7-34.7) for BNT162b2. Similar levels of added protective benefit, or 'boosting', was observed in very elderly and immunocompromised/high-risk individuals. Additional protection was highest within 2 months post-dosing, decreasing thereafter. Exploratory analyses revealed increased protection against severe in-hospital outcomes, including mortality. CONCLUSIONS Our results support the use of monovalent adenoviral/mRNA-based vaccine maintain protection against COVID-19 hospitalization from Omicron subvariants. However, optimal timing of booster vaccinations will need to be carefully considered for future booster strategies, especially among vulnerable subgroups.
Collapse
Affiliation(s)
- Wilhelmine Meeraus
- Medical Evidence, Vaccines & Immune Therapies, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK.
| | - Abigail Postema
- Formerly Medical Evidence, Vaccines & Immune Therapies, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | - Christen M Gray
- Real World Data Science, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | - Andrew Lee
- Medical and Payor Statistics, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | - Andre Santa Maria
- Early Phase Clinical Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Bárbara Emoingt Furtado
- Vaccines & Immune Therapies, BioPharmaceuticals Medical, AstraZeneca, Gaithersburg, Maryland, United States
| | - Eduardo Conde-Sousa
- Medical Evidence, Vaccines & Immune Therapies, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | - Mario Ouwens
- Medical and Payor Statistics, BioPharmaceuticals Medical, AstraZeneca, Gothenburg, Sweden
| | | | | | - Alexandre Naime Barbosa
- Department of Infectious Diseases, Botucatu School of Medicine, São Paulo State University, São Paulo, Brazil
| | - Claudia Corte
- Formerly Evidence Generation, AstraZeneca, São Paulo, Brazil
| | - Sylvia Taylor
- Medical Evidence, Vaccines & Immune Therapies, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| |
Collapse
|
2
|
Feldstein LR, Ruffin J, Wiegand RE, Borkowf CB, James-Gist J, Babu TM, Briggs-Hagen M, Chappell J, Chu HY, Englund JA, Kuntz JL, Lauring AS, Lo N, Carone M, Lockwood C, Martin ET, Midgley CM, Monto AS, Naleway AL, Ogilvie T, Saydah S, Schmidt MA, Schmitz JE, Smith N, Sohn I, Starita L, Talbot HK, Weil AA, Grijalva CG. Effectiveness of mRNA COVID-19 Vaccines and Hybrid Immunity in Preventing SARS-CoV-2 Infection and Symptomatic COVID-19 Among Adults in the United States. J Infect Dis 2025; 231:e743-e753. [PMID: 39774936 PMCID: PMC11998566 DOI: 10.1093/infdis/jiaf007] [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: 11/07/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Understanding protection against SARS-CoV-2 infection by vaccine and hybrid immunity is important for informing public health strategies as new variants emerge. METHODS We analyzed data from 3 cohort studies spanning 1 September 2022 to 31 July 2023 to estimate COVID-19 vaccine effectiveness (VE) against SARS-CoV-2 infection and symptomatic COVID-19 among adults with and without prior infection in the United States. Participants collected weekly nasal swabs irrespective of symptoms, participated in annual blood draws, and completed periodic surveys, which included vaccination status and infection history. Swabs were tested molecularly for SARS-CoV-2. VE was estimated by Cox proportional hazards models for the hazard ratios of infections, adjusting for covariates. VE was calculated considering prior infection and recency of vaccination. RESULTS Among 3344 adults, the adjusted VE of a bivalent vaccine against infection was 37.2% (95% CI, 12.3%-55.7%) within 7 to 59 days of vaccination and 21.1% (95% CI, -0.5% to 37.1%) within 60 to 179 days of vaccination when compared with participants who were unvaccinated or had received an original monovalent vaccine dose ≥180 days prior. Overall, the adjusted VE of a bivalent vaccine against infection, in conjunction with prior infection, was 62.2% (95% CI, 46.0%-74.5%) within 7 to 179 days of vaccination and 39.4% (95% CI, 12.5%-61.6%) at ≥180 days when compared with naive participants who were unvaccinated or had received a monovalent vaccine dose ≥180 days prior. CONCLUSIONS Adults with prior infection and recent vaccination had high protection against infection and symptomatic illness. Recent vaccination alone provided moderate protection.
Collapse
Affiliation(s)
- Leora R Feldstein
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jasmine Ruffin
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Ryan E Wiegand
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Craig B Borkowf
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jade James-Gist
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Tara M Babu
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington
| | - Melissa Briggs-Hagen
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - James Chappell
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Helen Y Chu
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington
| | - Janet A Englund
- Department of Pediatrics, University of Washington, Seattle Children's Research Institute, Seattle, Washington
| | | | - Adam S Lauring
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Natalie Lo
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington
| | - Marco Carone
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington
| | - Christina Lockwood
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington
| | - Emily T Martin
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Claire M Midgley
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Arnold S Monto
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | | | - Tara Ogilvie
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington
| | - Sharon Saydah
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Mark A Schmidt
- Kaiser Permanente Center for Health Research, Portland, Oregon
| | | | - Ning Smith
- Kaiser Permanente Center for Health Research, Portland, Oregon
| | - Ine Sohn
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lea Starita
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington
| | - H Keipp Talbot
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ana A Weil
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington
| | | |
Collapse
|
3
|
Hoeve CE, Neppelenbroek N, Vos ER, Huiberts AJ, Andeweg SP, den Hartog G, van Binnendijk R, de Melker H, van den Hof S, Knol M. Using SARS-CoV-2 nucleoprotein antibodies to detect (re)infection. Epidemiol Infect 2025; 153:e38. [PMID: 39915267 PMCID: PMC11869087 DOI: 10.1017/s095026882500010x] [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: 08/17/2024] [Revised: 11/28/2024] [Accepted: 01/16/2025] [Indexed: 03/01/2025] Open
Abstract
We assessed the validity of serum total anti-nucleoprotein Immunoglobulin (N-antibodies) to identify SARS-CoV-2 (re)infections by estimating the persistence of N-antibody seropositivity and boosting following infection. From a prospective Dutch cohort study (VASCO), we included adult participants with ≥2 consecutive self-collected serum samples, 4-8 months apart, between May 2021-May 2023. Sample pairs were stratified by N-seropositivity of the first sample and by self-reported infection within the sampling interval. We calculated the proportions of participants with N-seroconversion and fold-increase (1.5, 2, 3, 4) of N-antibody concentration over time since infection and explored determinants. We included 67,632 sample pairs. Pairs with a seronegative first sample (70%) showed 89% N-seroconversion after reported infection and 11% when no infection was reported. In pairs with a seropositive first sample (30%), 82%-65% showed a 1.5- to 4-fold increase with a reported reinfection, and 19%-10% without a reported reinfection, respectively. After one year, 83% remained N-seropositive post-first infection and 93%-61% showed a 1.5-fold to 4-fold increase post-reinfection. Odds for seroconversion/fold increase were higher for symptomatic infections and Omicron infections. In the current era with limited antigen or PCR testing, N-serology can be validly used to detect SARS-CoV-2 (re)infections at least up to a year after infection, supporting the monitoring of COVID-19 burden and vaccine effectiveness.
Collapse
Affiliation(s)
- Christel E. Hoeve
- National Institute for Public Health and the Environment, Center for Infectious Disease Control, Bilthoven, The Netherlands
| | - Nienke Neppelenbroek
- National Institute for Public Health and the Environment, Center for Infectious Disease Control, Bilthoven, The Netherlands
| | - Eric R.A. Vos
- National Institute for Public Health and the Environment, Center for Infectious Disease Control, Bilthoven, The Netherlands
| | - Anne J. Huiberts
- National Institute for Public Health and the Environment, Center for Infectious Disease Control, Bilthoven, The Netherlands
| | - Stijn P. Andeweg
- National Institute for Public Health and the Environment, Center for Infectious Disease Control, Bilthoven, The Netherlands
| | - Gerco den Hartog
- National Institute for Public Health and the Environment, Center for Infectious Disease Control, Bilthoven, The Netherlands
| | - Robert van Binnendijk
- National Institute for Public Health and the Environment, Center for Infectious Disease Control, Bilthoven, The Netherlands
| | - Hester de Melker
- National Institute for Public Health and the Environment, Center for Infectious Disease Control, Bilthoven, The Netherlands
| | - Susan van den Hof
- National Institute for Public Health and the Environment, Center for Infectious Disease Control, Bilthoven, The Netherlands
| | - Mirjam Knol
- National Institute for Public Health and the Environment, Center for Infectious Disease Control, Bilthoven, The Netherlands
| |
Collapse
|
4
|
Yan Y, Ito K, Fukuda H, Nojiri S, Urasaki W, Yamamoto T, Horiuchi Y, Hori S, Takahashi K, Naito T, Tabe Y. SARS-CoV-2 seroprevalence among healthcare workers in a highly vaccinated Japanese medical center from 2020-2023. Hum Vaccin Immunother 2024; 20:2337984. [PMID: 38622888 PMCID: PMC11027999 DOI: 10.1080/21645515.2024.2337984] [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: 12/06/2023] [Accepted: 03/28/2024] [Indexed: 04/17/2024] Open
Abstract
Infection-induced SARS-CoV-2 seroprevalence has been studied worldwide. At Juntendo University Hospital (JUH) in Tokyo, Japan, we have consistently conducted serological studies using the blood residue of healthcare workers (HCWs) at annual health examinations since 2020. In this 2023 study (n = 3,594), N-specific seroprevalence (infection-induced) was examined while univariate and multivariate logistic regression analyses were performed to compute ORs of seroprevalence with respect to basic characteristics of participants. We found that the N-specific seroprevalence in 2023 was 54.1%-a jump from 17.7% in 2022, and 1.6% in 2021-with 37.9% as non-PCR-confirmed asymptomatic infection cases. Those younger than 50 (adjusted OR = 1.62; p < .001) and recipients with 4 doses or less of vaccine had a higher risk to be N-positive, ranging from 1.45 times higher for the participants with 4 doses (p < .001) to 4.31 times higher for the participants with 1 dose (p < .001), compared to those with 5 or more doses. Our findings indicate that robust vaccination programs may have helped alleviate symptoms but consequently caused asymptomatic spread in this hospital, especially among younger HCWs. Although having four doses or less was found to be associated with higher risk of infection, the optimal constitution and intervals for effective booster vaccines warrant further investigations.
Collapse
Affiliation(s)
- Yan Yan
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Kanami Ito
- Department of Safety and Health Promotion, Juntendo University, Tokyo, Japan
| | - Hiroshi Fukuda
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Safety and Health Promotion, Juntendo University, Tokyo, Japan
| | - Shuko Nojiri
- Medical Technology Innovation Center, Juntendo University, Tokyo, Japan
| | - Wataru Urasaki
- Clinical Research and Trial Center, Juntendo University, Tokyo, Japan
- Graduate School of Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Takamasa Yamamoto
- Department of Clinical Laboratory, Juntendo University Hospital, Tokyo, Japan
| | - Yuki Horiuchi
- Department of Clinical Laboratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Satoshi Hori
- Infection Control Science, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Toshio Naito
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Safety and Health Promotion, Juntendo University, Tokyo, Japan
| | - Yoko Tabe
- Department of Clinical Laboratory, Juntendo University Hospital, Tokyo, Japan
- Department of Clinical Laboratory Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Fernández-Ciriza L, del Pozo JL, Betanzos N, González Á, Fernandez-Montero A, Carmona-Torre F, Vidaurreta M, Carlos S, Reina G. Impact of Repeated Variant Exposures on Cellular and Humoral Immunogenicity Induced by SARS-CoV-2 Vaccines. Vaccines (Basel) 2024; 12:1408. [PMID: 39772069 PMCID: PMC11680034 DOI: 10.3390/vaccines12121408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES The emergence of the Omicron variant has complicated COVID-19 control and prompted vaccine updates. Recent studies have shown that a fourth dose significantly protects against infection and severe disease, though long-term immunity data remain limited. This study aimed to assess Anti-S-RBD antibodies and interferon-γ levels in healthcare workers 12 months after receiving bivalent Original/Omicron BA.4-5 fourth SARS-CoV-2 vaccine. METHODS In this prospective cohort study, 549 healthcare workers were categorized by the initial vaccination schedule, with 229 individuals having received the fourth SARS-CoV-2 vaccine dose. Blood samples were collected from all participants 12 months post-vaccination. RESULTS Significant differences in Anti-S-RBD antibody levels were observed between those receiving a fourth dose and those who did not, while no differences were seen in interferon-γ levels. After 12 months, there were no significant differences in humoral and cellular immunity response between volunteers primoinfected or reinfected across different periods by the Omicron variant. A multivariable analysis revealed an association between high antibody levels (>6000 U/mL) and interferon-γ levels (OR: 3.13; 95% CI: 1.3-7.7; p < 0.05). Regarding primary vaccine schedules, participants vaccinated with ChAdOx1 (a single or double dose) had notably lower antibody levels compared to those who received mRNA-based vaccines. Additionally, the study shows a higher frequency of multiple infections among those with a single-dose ChAdOx1 primary schedule (OR: 6.24; 95% CI: 1.25-31.15; p < 0.01). CONCLUSIONS Overall, mRNA-based vaccines exhibited stronger long-term immunogenicity. Repeated exposure to the Omicron variant seems to mitigate immune imprinting from the wild-type SARS-CoV-2. An association was observed between high antibody levels and a strong cellular response, although the correlation was not linear.
Collapse
Affiliation(s)
- Leire Fernández-Ciriza
- Department of Microbiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (L.F.-C.); (N.B.); (M.V.); (G.R.)
| | - José Luis del Pozo
- Department of Microbiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (L.F.-C.); (N.B.); (M.V.); (G.R.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
- Infectious Diseases Division, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Nazaret Betanzos
- Department of Microbiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (L.F.-C.); (N.B.); (M.V.); (G.R.)
| | - Álvaro González
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
- Department of Biochemistry, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Alejandro Fernandez-Montero
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
- Department of Occupational Medicine, Universidad de Navarra, 31008 Pamplona, Spain
| | - Francisco Carmona-Torre
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
- Infectious Diseases Division, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Marta Vidaurreta
- Department of Microbiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (L.F.-C.); (N.B.); (M.V.); (G.R.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
- Department of Biochemistry, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Silvia Carlos
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
- Department of Preventive Medicine and Public Health, Universidad de Navarra, 31008 Pamplona, Spain
| | - Gabriel Reina
- Department of Microbiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (L.F.-C.); (N.B.); (M.V.); (G.R.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
| |
Collapse
|
6
|
Ciesla A, Mak J, Roper L, Fleming‐Dutra K, Smith Z, Wiegand R, Britton A, Miller J, Link‐Gelles R. Effectiveness of Bivalent mRNA Vaccines in Preventing Symptomatic SARS-CoV-2 Infection-Increasing Community Access to Testing Program, United States, January-September 2023. Influenza Other Respir Viruses 2024; 18:e70038. [PMID: 39522959 PMCID: PMC11550894 DOI: 10.1111/irv.70038] [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: 08/05/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND On September 2, 2022, bivalent COVID-19 mRNA vaccines, were recommended to address reduced effectiveness of COVID-19 monovalent vaccines during SARS-CoV-2 Omicron variant predominance. METHODS Using national pharmacy-based SARS-CoV-2 testing program data from January 15 to September 11, 2023, this test-negative, case-control design study assessed bivalent COVID-19 vaccine effectiveness (VE) against symptomatic infection. RESULTS VE against symptomatic infection of a bivalent dose between 2 weeks and 1 month after bivalent vaccination ranged from 46% (95% CI: 38%-52%) for those aged ≥ 65 years to 61% (95% CI 41%-75%) for those aged 12-17 years. CONCLUSION Bivalent vaccines protected against symptomatic infection. However, effectiveness waned over time, emphasizing the need to stay up to date with COVID-19 vaccination.
Collapse
Affiliation(s)
- Allison Avrich Ciesla
- National Center for Immunization and Respiratory DiseasesCenters for Disease Control and PreventionAtlantaGeorgiaUSA
- Eagle Health AnalyticsSan AntonioTexasUSA
| | - Josephine Mak
- National Center for Immunization and Respiratory DiseasesCenters for Disease Control and PreventionAtlantaGeorgiaUSA
| | - Lauren E. Roper
- National Center for Immunization and Respiratory DiseasesCenters for Disease Control and PreventionAtlantaGeorgiaUSA
| | - Katherine E. Fleming‐Dutra
- National Center for Immunization and Respiratory DiseasesCenters for Disease Control and PreventionAtlantaGeorgiaUSA
| | - Zachary R. Smith
- Office of Readiness and ResponseCenters for Disease Control and PreventionAtlantaGeorgiaUSA
| | - Ryan E. Wiegand
- National Center for Immunization and Respiratory DiseasesCenters for Disease Control and PreventionAtlantaGeorgiaUSA
| | - Amadea Britton
- National Center for Immunization and Respiratory DiseasesCenters for Disease Control and PreventionAtlantaGeorgiaUSA
| | - Joseph Miller
- Office of Readiness and ResponseCenters for Disease Control and PreventionAtlantaGeorgiaUSA
| | - Ruth Link‐Gelles
- National Center for Immunization and Respiratory DiseasesCenters for Disease Control and PreventionAtlantaGeorgiaUSA
- US Public Health Service Commissioned CorpsRockvilleMarylandUSA
| |
Collapse
|
7
|
Huiberts AJ, Hoeve CE, Kooijman MN, de Melker HE, Hahné SJM, Grobbee DE, van Binnendijk R, den Hartog G, van de Wijgert JHHM, van den Hof S, Knol MJ. Cohort profile: an observational population-based cohort study on COVID-19 vaccine effectiveness in the Netherlands - the VAccine Study COVID-19 (VASCO). BMJ Open 2024; 14:e085388. [PMID: 39401962 PMCID: PMC11474871 DOI: 10.1136/bmjopen-2024-085388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/20/2024] [Indexed: 10/17/2024] Open
Abstract
PURPOSE VAccine Study COVID-19 (VASCO) is a cohort study with a 5-year follow-up that was initiated when COVID-19 vaccination was introduced in the Netherlands. The primary objective is to estimate real-world vaccine effectiveness (VE) of COVID-19 vaccines against SARS-CoV-2 infection in the Netherlands, overall and in four subpopulations defined by age and medical risk. PARTICIPANTS The cohort consists of 45 547 community-dwelling participants aged 18-85 years who were included irrespective of their COVID-19 vaccination status or intention to get vaccinated. A medical risk condition is present in 4289 (19.8%) of 21 679 individuals aged 18-59 years, and in 9135 (38.3%) of 23 821 individuals aged 60-85 years. After 1 year of follow-up, 5502 participants had dropped out of the study. At inclusion and several times after inclusion, participants are asked to take a self-collected fingerprick blood sample in which nucleoprotein and spike protein receptor binding domain-specific antibody concentrations are assessed. Participants are also asked to complete monthly digital questionnaires in the first year, and 3 monthly in years 2-5, including questions on sociodemographic factors, health status, COVID-19 vaccination, SARS-CoV-2-related symptoms and testing results, and behavioural responses to COVID-19 measures. FINDINGS TO DATE VASCO data have been used to describe VE against SARS-CoV-2 infection of primary vaccination, first and second booster and bivalent boosters, the impact of hybrid immunity on SARS-CoV-2 infection and VE against infectiousness. Furthermore, data were used to describe antibody response following vaccination and breakthrough infections and to investigate the relation between antibody response and reactogenicity. FUTURE PLANS VASCO will be able to contribute to policy decision-making regarding future COVID-19 vaccination. Furthermore, VASCO provides an infrastructure to conduct further studies and to respond to changes in vaccination campaigns and testing policy, and new virus variants. TRIAL REGISTRATION NUMBER NL9279.
Collapse
Affiliation(s)
- Anne J Huiberts
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Christina E Hoeve
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Marjolein N Kooijman
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Hester E de Melker
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Susan JM Hahné
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Diederick E Grobbee
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Rob van Binnendijk
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Gerco den Hartog
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Janneke HHM van de Wijgert
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Susan van den Hof
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Mirjam J Knol
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| |
Collapse
|
8
|
Bolormaa E, Shim J, Choi YS, Kwon D, Choe YJ, Choe SA. Methodology of comparative studies on the relative effectiveness of COVID-19 vaccines: a systematic review. Osong Public Health Res Perspect 2024; 15:395-408. [PMID: 39511961 PMCID: PMC11563719 DOI: 10.24171/j.phrp.2024.0063] [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: 03/09/2024] [Revised: 07/30/2024] [Accepted: 08/26/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND This study aimed to comprehensively outline the methodological approaches used in published research comparing the vaccine effectiveness (VE) of coronavirus disease 2019 (COVID-19) vaccines. METHODS A systematic search was conducted on June 13, 2024, to identify comparative studies evaluating the effectiveness of mRNA versus non-mRNA and monovalent versus bivalent COVID-19 vaccines. We screened titles, abstracts, and full texts, collecting data on publication year, country, sample size, study population composition, study design, VE estimates, outcomes, and covariates. Studies that reported relative VE (rVE) were analyzed separately from those that did not. RESULTS We identified 25 articles comparing rVE between mRNA and non-mRNA COVID-19 vaccines, as well as between monovalent and bivalent formulations. Among the studies assessing VE by vaccine type, 126 did not provide rVE estimates. Comparative VE studies frequently employed retrospective cohort designs. Among the definitions of rVE used, the most common were hazard ratio and absolute VE, calculated as (1-odds ratio)×100. Studies were most frequently conducted in the United Kingdom and the United States, and the most common outcome was infection. Most targeted the general population and assessed the VE of mRNA vaccines using the AstraZeneca vaccine as a reference. A small proportion, 7.3% (n=11), did not adjust for any variables. Only 3 studies (2.0%) adjusted for all core confounding variables recommended by the World Health Organization. CONCLUSION Few comparative studies of COVID-19 vaccines have incorporated rVE methodologies. Reporting rVE and employing a consistent set of covariates can broaden our understanding of COVID-19 vaccines.
Collapse
Affiliation(s)
- Erdenetuya Bolormaa
- Department of Preventive Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jiae Shim
- Division of Epidemiological Investigation Analysis, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Young-Sook Choi
- Division of Epidemiological Investigation Analysis, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Donghyok Kwon
- Division of Epidemiological Investigation Analysis, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Young June Choe
- Department of Pediatrics, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seung-Ah Choe
- Department of Preventive Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
9
|
Kanokudom S, Chansaenroj J, Suntronwong N, Wongsrisang L, Aeemjinda R, Vichaiwattana P, Thatsanathorn T, Chantima W, Pakchotanon P, Duangchinda T, Sudhinaraset N, Honsawek S, Poovorawan Y. Safety and antibody responses of Omicron BA.4/5 bivalent booster vaccine among hybrid immunity with diverse vaccination histories: A cohort study. Vaccine X 2024; 20:100538. [PMID: 39211731 PMCID: PMC11359987 DOI: 10.1016/j.jvacx.2024.100538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 09/04/2024] Open
Abstract
This cohort study, conducted between July and August 2023, evaluated the adverse events (AEs) and immune response to a bivalent mRNA-1273.222 (containing sequences of the original Wuhan-H1 strain and the Omicron BA.4/5 variant) booster vaccine in 122 participants. The study included individuals with diverse vaccination histories, and their responses were assessed based on anti-receptor binding domain (RBD) IgG levels and neutralizing antibodies against the wild-type, Omicron BA.5, and XBB.1.16 variants. Following administration of the BA.4/5 bivalent vaccine, AEs were generally mild to moderate and well-tolerated within a few days. There were no reports of vomiting and no serious AEs or death. The findings demonstrated robust immune responses, with significant increases in anti-RBD IgG levels, particularly in groups that had received 3 -6 doses before the booster dose. The BA.4/5 bivalent booster effectively induced neutralizing antibodies against the vaccine strains, providing robust neutralization, including the XBB.1.16 strain. The study also highlighted that individuals with hybrid immunity, especially those assumed infected with the BA.5 strain or who had been infected twice, showed higher levels of robust neutralizing activity against Omicron XBB.1.16. Overall, these results indicate that the BA.4/5 bivalent booster vaccines can induce potent and good antibody responses in emerging Omicron subvariants, supporting its efficacy as a booster in individuals with diverse vaccination histories.
Collapse
Affiliation(s)
- Sitthichai Kanokudom
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Osteoarthritis and Musculoskeleton, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Jira Chansaenroj
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nungruthai Suntronwong
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Lakkhana Wongsrisang
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Ratchadawan Aeemjinda
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Preeyaporn Vichaiwattana
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Thaksaporn Thatsanathorn
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Warangkana Chantima
- Division of Dengue Hemorrhagic Fever Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Pattarakul Pakchotanon
- Molecular Biology of Dengue and Flaviviruses Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Thaneeya Duangchinda
- Molecular Biology of Dengue and Flaviviruses Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Natthinee Sudhinaraset
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sittisak Honsawek
- Center of Excellence in Osteoarthritis and Musculoskeleton, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Fellow of the Royal Society of Thailand (FRS [T]), The Royal Society of Thailand, Sanam Sueapa, Dusit, Bangkok 10300, Thailand
| |
Collapse
|
10
|
Gelderloos AT, Verheul MK, Middelhof I, de Zeeuw-Brouwer ML, van Binnendijk RS, Buisman AM, van Kasteren PB. Repeated COVID-19 mRNA vaccination results in IgG4 class switching and decreased NK cell activation by S1-specific antibodies in older adults. Immun Ageing 2024; 21:63. [PMID: 39272189 PMCID: PMC11401348 DOI: 10.1186/s12979-024-00466-9] [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: 07/17/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Previous research has shown that repeated COVID-19 mRNA vaccination leads to a marked increase of SARS-CoV-2 spike-specific serum antibodies of the IgG4 subclass, indicating far-reaching immunoglobulin class switching after booster immunization. Considering that repeated vaccination has been recommended especially for older adults, the aim of this study was to investigate IgG subclass responses in the ageing population and assess their relation with Fc-mediated antibody effector functionality. RESULTS Spike S1-specific IgG subclass concentrations (expressed in arbitrary units per mL), antibody-dependent NK cell activation, complement deposition and monocyte phagocytosis were quantified in serum from older adults (n = 38-50, 65-83 years) at one month post-second, -third and -fifth vaccination. Subclass distribution in serum was compared to that in younger adults (n = 64, 18-47 years) at one month post-second and -third vaccination. Compared to younger individuals, older adults showed increased levels of IgG2 and IgG4 at one month post-third vaccination (possibly related to factors other than age) and a further increase following a fifth dose. The capacity of specific serum antibodies to mediate NK cell activation and complement deposition relative to S1-specific total IgG concentrations decreased upon repeated vaccination. This decrease associated with an increased IgG4/IgG1 ratio. CONCLUSIONS In conclusion, these findings show that, like younger individuals, older adults produce antibodies with reduced functional capacity upon repeated COVID-19 mRNA vaccination. Additional research is needed to better understand the mechanisms underlying these responses and their potential implications for vaccine effectiveness. Such knowledge is vital for the future design of optimal vaccination strategies in the ageing population.
Collapse
Affiliation(s)
- Anne T Gelderloos
- Center for Immunology of Infectious Diseases and Vaccines (IIV), Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Marije K Verheul
- Center for Immunology of Infectious Diseases and Vaccines (IIV), Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Irene Middelhof
- Center for Immunology of Infectious Diseases and Vaccines (IIV), Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Mary-Lène de Zeeuw-Brouwer
- Center for Immunology of Infectious Diseases and Vaccines (IIV), Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Robert S van Binnendijk
- Center for Immunology of Infectious Diseases and Vaccines (IIV), Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Anne-Marie Buisman
- Center for Immunology of Infectious Diseases and Vaccines (IIV), Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Puck B van Kasteren
- Center for Immunology of Infectious Diseases and Vaccines (IIV), Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands.
| |
Collapse
|
11
|
Costa Clemens SA, Jepson B, Bhorat QE, Ahmad A, Akhund T, Aley PK, Bansal H, Bibi S, Kelly EJ, Khan M, Lambe T, Lombaard JJ, Matthews S, Pipolo Milan E, Olsson U, Ramasamy MN, Moura de Oliveira Paiva MS, Seegobin S, Shoemaker K, Szylak A, Villafana T, Pollard AJ, Green JA. Immunogenicity and safety of beta variant COVID-19 vaccine AZD2816 and AZD1222 (ChAdOx1 nCoV-19) as primary-series vaccination for previously unvaccinated adults in Brazil, South Africa, Poland, and the UK: a randomised, partly double-blinded, phase 2/3 non-inferiority immunobridging study. THE LANCET. MICROBE 2024; 5:100863. [PMID: 38878794 DOI: 10.1016/s2666-5247(24)00078-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/12/2024] [Accepted: 03/12/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND AZD2816 is a variant-adapted COVID-19 vaccine that expresses the full-length SARS-CoV-2 beta variant spike protein but is otherwise similar to AZD1222 (ChAdOx1 nCoV-19). This study aimed to evaluate the safety and immunogenicity of AZD1222 or AZD2816 (or both) primary-series vaccination in a cohort of adult participants who were previously unvaccinated. METHODS In this phase 2/3, randomised, multinational, active-controlled, non-inferiority, immunobridging study, adult participants previously unvaccinated for COVID-19 were enrolled at 16 study sites in Brazil, South Africa, Poland, and the UK. Participants were stratified by age, sex, and comorbidity and randomly assigned 5:5:5:2 to receive a primary series of AZD1222 (AZD1222 group), AZD2816 (AZD2816 [4-week] group), or AZD1222-AZD2816 (AZD1222-AZD2816 group) at 4-week dosing intervals, or AZD2816 at a 12-week interval (AZD2816 [12-week] group) and evaluated for safety and immunogenicity through 180 days after dose 2. Primary outcomes were safety (rates of solicited adverse events occurring during 7 days and unsolicited adverse events occurring during 28 days after each dose) and immunogenicity (non-inferiority of pseudovirus neutralising antibody geometric mean titre [GMT], GMT ratio margin of 0·67, and seroresponse rate, rate difference margin of -10%, recorded 28 days after dose 2 with AZD2816 [4-week interval] against beta vs AZD1222 against ancestral SARS-CoV-2) in participants who were seronegative at baseline. This trial is registered with ClinicalTrials.gov, NCT04973449, and is completed. FINDINGS Between July 7 and Nov 12, 2021, 1449 participants were assigned to the AZD1222 group (n=413), the AZD2816 (4-week) group (n=415), the AZD1222-AZD2816 group (n=412), and the AZD2816 (12-week) group (n=209). Ten (2·6%) of 378 participants who were seronegative at baseline in the AZD1222 group, nine (2·4%) of 379 in the AZD2816 (4-week) group, eight (2·1%) of 380 in the AZD1222-AZD2816 group, and 11 (5·8%) of 191 in the AZD2816 (12-week) group had vaccine-related unsolicited adverse events. Serious adverse events were recorded in one (0·3%) participant in the AZD1222 group, one (0·3%) in the AZD2816 (4-week) group, two (0·5%) in the AZD1222-AZD2816 group, and none in the AZD2816 (12-week) group. Co-primary immunogenicity endpoints were met: neutralising antibody GMT (ratio 1·19 [95% CI 1·08-1·32]; lower bound greater than 0·67) and seroresponse rate (difference 1·7% [-3·1 to 6·5]; lower bound greater than -10%) at 28 days after dose 2 were non-inferior in the AZD2816 (4-week) group against beta versus in the AZD1222 group against ancestral SARS-CoV-2. Seroresponse rates were highest with AZD2816 against beta (12-week interval 94·3% [95% CI 89·4-97·3]; 4-week interval 85·7% [81·5-89·2]) and with AZD1222 (84·6% [80·3-88·2]) against ancestral SARS-CoV-2. INTERPRETATION Primary series of AZD1222 and AZD2816 were well tolerated, with no emergent safety concerns. Both vaccines elicited robust immunogenicity against beta and ancestral SARS-CoV-2 with greater responses demonstrated when testing against SARS-CoV-2 strains that matched those targeted by the respective vaccine. These findings demonstrate the continued importance of ancestral COVID-19 vaccines in protecting against severe COVID-19 and highlight the feasibility of using the ChAdOx1 platform to develop COVID-19 vaccines against future SARS-CoV-2 variants. FUNDING AstraZeneca.
Collapse
Affiliation(s)
- Sue Ann Costa Clemens
- Department of Paediatrics, University of Oxford, Oxford, UK; Institute for Global Health, Siena University, Siena, Italy
| | - Brett Jepson
- Biometrics, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Qasim E Bhorat
- Soweto Clinical Trials Centre, Soweto, Gauteng, South Africa
| | - Abdullahi Ahmad
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Tauseefullah Akhund
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Parvinder K Aley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Himanshu Bansal
- Biometrics, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Elizabeth J Kelly
- Formerly Translational Medicine, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Mark Khan
- Clinical Development, BioPharmaceuticals R&D, AstraZeneca, Mississauga, ON, Canada
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Chinese Academy of Medical Science, Oxford Institute, University of Oxford, Oxford, UK
| | | | - Sam Matthews
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Eveline Pipolo Milan
- Centro de Estudos e Pesquisas em Moléstias Infec, Centro de Pesquisas Clínicas de Natal, Natal, Rio Grande do Norte, Brazil
| | - Urban Olsson
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maheshi N Ramasamy
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Seth Seegobin
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Kathryn Shoemaker
- Biometrics, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ameena Szylak
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Mississauga, ON, Canada
| | - Tonya Villafana
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Justin A Green
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
12
|
Song S, Madewell ZJ, Liu M, Miao Y, Xiang S, Huo Y, Sarkar S, Chowdhury A, Longini IM, Yang Y. A systematic review and meta-analysis on the effectiveness of bivalent mRNA booster vaccines against Omicron variants. Vaccine 2024; 42:3389-3396. [PMID: 38653679 DOI: 10.1016/j.vaccine.2024.04.049] [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: 01/22/2024] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND A global shift to bivalent mRNA vaccines is ongoing to counterbalance the diminishing effectiveness of the original monovalent vaccines due to the evolution of SARS-CoV-2 variants, yet substantial variation in the bivalent vaccine effectiveness (VE) exists across studies and a complete picture is lacking. METHODS We searched papers evaluating absolute or relative effectiveness of SARS-CoV-2 BA.1 type or BA.4/5 type bivalent mRNA vaccines on eight publication databases published from September 1st, 2022, to November 8th, 2023. Pooled VE against Omicron-associated infection and severe events (hospitalization and/or death) was estimated in reference to unvaccinated, ≥2 original monovalent doses, and ≥ 3 original monovalent doses. RESULTS From 630 citations identified, 28 studies were included, involving 55,393,303 individuals. Bivalent boosters demonstrated higher effectiveness against symptomatic or any infection for all ages combined, with an absolute VE of 53.5 % (95 % CI: -22.2-82.3 %) when compared to unvaccinated and relative VE of 30.8 % (95 % CI: 22.5-38.2 %) and 28.4 % (95 % CI: 10.2-42.9 %) when compared to ≥ 2 and ≥ 3 original monovalent doses, respectively. The corresponding VE estimates for adults ≥ 60 years old were 22.5 % (95 % CI: 16.8-39.8 %), 31.4 % (95 % CI: 27.7-35.0 %), and 30.6 % (95 % CI: -13.2-57.5 %). Pooled bivalent VE estimates against severe events were higher, 72.9 % (95 % CI: 60.5-82.4 %), 57.6 % (95 % CI: 42.4-68.8 %), and 62.1 % (95 % CI: 54.6-68.3 %) for all ages, and 72.0 % (95 % CI: 51.4-83.9 %), 63.4 % (95 % CI: 41.0-77.3 %), and 60.7 % (95 % CI: 52.4-67.6 %) for adults ≥ 60 years old, compared to unvaccinated, ≥2 original monovalent doses, and ≥ 3 original monovalent doses, respectively. CONCLUSIONS The bivalent boosters demonstrated superior protection against severe outcomes than the original monovalent boosters across age groups, highlighting the critical need for improving vaccine coverage, especially among the vulnerable older subpopulation.
Collapse
Affiliation(s)
- Shangchen Song
- Department of Biostatistics, College of Public Health and health Professions, University of Florida, Gainesville, FL, USA
| | - Zachary J Madewell
- Department of Biostatistics, College of Public Health and health Professions, University of Florida, Gainesville, FL, USA
| | - Mingjin Liu
- Department of Statistics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Yu Miao
- Department of Statistics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Shaolin Xiang
- Department of Statistics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Yanan Huo
- Gilead Sciences, Inc, Foster City, CA, USA
| | - Shoumi Sarkar
- Department of Biostatistics, College of Public Health and health Professions, University of Florida, Gainesville, FL, USA
| | - Amily Chowdhury
- Department of Computer Science, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Ira M Longini
- Department of Biostatistics, College of Public Health and health Professions, University of Florida, Gainesville, FL, USA
| | - Yang Yang
- Department of Statistics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA.
| |
Collapse
|
13
|
Plumb ID, Briggs Hagen M, Wiegand R, Dumyati G, Myers C, Harland KK, Krishnadasan A, James Gist J, Abedi G, Fleming-Dutra KE, Chea N, Lee JE, Kellogg M, Edmundson A, Britton A, Wilson LE, Lovett SA, Ocampo V, Markus TM, Smithline HA, Hou PC, Lee LC, Mower W, Rwamwejo F, Steele MT, Lim SC, Schrading WA, Chinnock B, Beiser DG, Faine B, Haran JP, Nandi U, Chipman AK, LoVecchio F, Eucker S, Femling J, Fuller M, Rothman RE, Curlin ME, Talan DA, Mohr NM. Effectiveness of a bivalent mRNA vaccine dose against symptomatic SARS-CoV-2 infection among U.S. Healthcare personnel, September 2022-May 2023. Vaccine 2024; 42:2543-2552. [PMID: 37973512 PMCID: PMC10994739 DOI: 10.1016/j.vaccine.2023.10.072] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Bivalent mRNA vaccines were recommended since September 2022. However, coverage with a recent vaccine dose has been limited, and there are few robust estimates of bivalent VE against symptomatic SARS-CoV-2 infection (COVID-19). We estimated VE of a bivalent mRNA vaccine dose against COVID-19 among eligible U.S. healthcare personnel who had previously received monovalent mRNA vaccine doses. METHODS We conducted a case-control study in 22 U.S. states, and enrolled healthcare personnel with COVID-19 (case-participants) or without COVID-19 (control-participants) during September 2022-May 2023. Participants were considered eligible for a bivalent mRNA dose if they had received 2-4 monovalent (ancestral-strain) mRNA vaccine doses, and were ≥67 days after the most recent vaccine dose. We estimated VE of a bivalent mRNA dose using conditional logistic regression, accounting for matching by region and four-week calendar period. We adjusted estimates for age group, sex, race and ethnicity, educational level, underlying health conditions, community COVID-19 exposure, prior SARS-CoV-2 infection, and days since the last monovalent mRNA dose. RESULTS Among 3,647 healthcare personnel, 1,528 were included as case-participants and 2,119 as control-participants. Participants received their last monovalent mRNA dose a median of 404 days previously; 1,234 (33.8%) also received a bivalent mRNA dose a median of 93 days previously. Overall, VE of a bivalent dose was 34.1% (95% CI, 22.6%-43.9%) against COVID-19 and was similar by product, days since last monovalent dose, number of prior doses, age group, and presence of underlying health conditions. However, VE declined from 54.8% (95% CI, 40.7%-65.6%) after 7-59 days to 21.6% (95% CI 5.6%-34.9%) after ≥60 days. CONCLUSIONS Bivalent mRNA COVID-19 vaccines initially conferred approximately 55% protection against COVID-19 among U.S. healthcare personnel. However, protection waned after two months. These findings indicate moderate initial protection against symptomatic SARS-CoV-2 infection by remaining up-to-date with COVID-19 vaccines.
Collapse
Affiliation(s)
- Ian D Plumb
- National Center for Immunizations and Respiratory Diseases, Centers for Disease Control & Prevention, Atlanta, GA, USA.
| | - Melissa Briggs Hagen
- National Center for Immunizations and Respiratory Diseases, Centers for Disease Control & Prevention, Atlanta, GA, USA
| | - Ryan Wiegand
- National Center for Immunizations and Respiratory Diseases, Centers for Disease Control & Prevention, Atlanta, GA, USA
| | - Ghinwa Dumyati
- University of Rochester Medical Center, Rochester, NY, USA
| | | | | | | | - Jade James Gist
- National Center for Immunizations and Respiratory Diseases, Centers for Disease Control & Prevention, Atlanta, GA, USA
| | - Glen Abedi
- National Center for Immunizations and Respiratory Diseases, Centers for Disease Control & Prevention, Atlanta, GA, USA
| | - Katherine E Fleming-Dutra
- National Center for Immunizations and Respiratory Diseases, Centers for Disease Control & Prevention, Atlanta, GA, USA
| | - Nora Chea
- National Center for Emerging and Zoonotic Diseases, Centers for Disease Control & Prevention, USA
| | - Jane E Lee
- California Emerging Infections Program, Oakland, CA, USA
| | | | - Alexandra Edmundson
- Connecticut Emerging Infections Program, Yale School of Public Health, CT, USA
| | - Amber Britton
- Georgia Emerging Infections Program and Emory University School of Medicine, Atlanta, GA, USA
| | - Lucy E Wilson
- Maryland Emerging Infections Program, Maryland Department of Health and University of Maryland, Baltimore, MD, USA
| | | | - Valerie Ocampo
- Public Health Division, Oregon Health Authority, OR, USA
| | | | | | - Peter C Hou
- Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | - Mark T Steele
- University of Missouri-Kansas City, Kansas City, MO, USA
| | - Stephen C Lim
- University Medical Center New Orleans, LSU Health Sciences Center, New Orleans, LA, USA
| | | | | | | | | | - John P Haran
- University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Utsav Nandi
- University of Mississippi Medical Center, Jackson, MS, USA
| | | | | | | | - Jon Femling
- University of New Mexico Health Science Center, USA
| | | | - Richard E Rothman
- Department of Emergency Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | | | | |
Collapse
|
14
|
Liu R, Natekar JP, Kim KH, Pathak H, Bhatnagar N, Raha JR, Park BR, Guglani A, Shin CH, Kumar M, Kang SM. Multivalent and Sequential Heterologous Spike Protein Vaccinations Effectively Induce Protective Humoral Immunity against SARS-CoV-2 Variants. Vaccines (Basel) 2024; 12:362. [PMID: 38675744 PMCID: PMC11053539 DOI: 10.3390/vaccines12040362] [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: 03/06/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
The emergence of new SARS-CoV-2 variants continues to cause challenging problems for the effective control of COVID-19. In this study, we tested the hypothesis of whether a strategy of multivalent and sequential heterologous spike protein vaccinations would induce a broader range and higher levels of neutralizing antibodies against SARS-CoV-2 variants and more effective protection than homologous spike protein vaccination in a mouse model. We determined spike-specific IgG, receptor-binding inhibition titers, and protective efficacy in the groups of mice that were vaccinated with multivalent recombinant spike proteins (Wuhan, Delta, Omicron), sequentially with heterologous spike protein variants, or with homologous spike proteins. Trivalent (Wuhan + Delta + Omicron) and sequential heterologous spike protein vaccinations were more effective in inducing serum inhibition activities of receptor binding to spike variants and virus neutralizing antibody titers than homologous spike protein vaccination. The higher efficacy of protection was observed in mice with trivalent and sequential heterologous spike protein vaccination after a challenge with a mouse-adapted SARS-CoV-2 MA10 strain compared to homologous spike protein vaccination. This study provides evidence that a strategy of multivalent and sequential heterologous variant spike vaccination might provide more effective protection against emerging SARS-CoV-2 variants than homologous spike vaccination and significantly alleviate severe inflammation due to COVID-19.
Collapse
Affiliation(s)
- Rong Liu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Janhavi P. Natekar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (J.P.N.); (H.P.)
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Heather Pathak
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (J.P.N.); (H.P.)
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Jannatul Ruhan Raha
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Bo Ryoung Park
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Anchala Guglani
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (J.P.N.); (H.P.)
| | - Chong Hyun Shin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (J.P.N.); (H.P.)
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (R.L.); (K.-H.K.); (N.B.); (J.R.R.); (B.R.P.); (C.H.S.)
| |
Collapse
|
15
|
Sop J, Traut CC, Dykema AG, Hunt JH, Beckey TP, Basseth CR, Antar AAR, Laeyendecker O, Smith KN, Blankson JN. Bivalent mRNA COVID vaccines elicit predominantly cross-reactive CD4 + T cell clonotypes. Cell Rep Med 2024; 5:101442. [PMID: 38423018 PMCID: PMC10983033 DOI: 10.1016/j.xcrm.2024.101442] [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: 09/21/2023] [Revised: 12/28/2023] [Accepted: 02/03/2024] [Indexed: 03/02/2024]
Abstract
Bivalent COVID vaccines containing mRNA for ancestral and Omicron BA.5 spike proteins do not induce stronger T cell responses to Omicron BA.5 spike proteins than monovalent vaccines that contain only ancestral spike mRNA. The reasons for this finding have not been elucidated. Here, we show that healthy donors (HDs) and people living with HIV (PLWH) on antiretroviral therapy mostly target T cell epitopes that are not affected by BA.5 mutations. We use the functional expansion of specific T cells (FEST) assay to determine the percentage of CD4+ T cells that cross-recognize both spike proteins and those that are monoreactive for each protein. We show a predominance of cross-reactive CD4+ T cells; less than 10% percent of spike-specific CD4+ T cell receptors were BA.5 monoreactive in most HDs and PLWH. Our data suggest that the current bivalent vaccines do not induce robust BA.5-monoreactive T cell responses.
Collapse
Affiliation(s)
- Joel Sop
- Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Caroline C Traut
- Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Arbor G Dykema
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Joanne H Hunt
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Baltimore, MD, USA
| | - Tyler P Beckey
- Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA
| | | | | | - Oliver Laeyendecker
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Baltimore, MD, USA
| | - Kellie N Smith
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
| | - Joel N Blankson
- Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA.
| |
Collapse
|
16
|
Huiberts AJ, Hoeve CE, de Gier B, Cremer J, van der Veer B, de Melker HE, van de Wijgert JH, van den Hof S, Eggink D, Knol MJ. Effectiveness of Omicron XBB.1.5 vaccine against infection with SARS-CoV-2 Omicron XBB and JN.1 variants, prospective cohort study, the Netherlands, October 2023 to January 2024. Euro Surveill 2024; 29:2400109. [PMID: 38456217 PMCID: PMC10986669 DOI: 10.2807/1560-7917.es.2024.29.10.2400109] [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: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024] Open
Abstract
We estimated vaccine effectiveness (VE) of SARS-CoV-2 Omicron XBB.1.5 vaccination against self-reported infection between 9 October 2023 and 9 January 2024 in 23,895 XBB.1.5 vaccine-eligible adults who had previously received at least one booster. VE was 41% (95% CI: 23-55) in 18-59-year-olds and 50% (95% CI: 44-56) in 60-85-year-olds. Sequencing data suggest lower protection against the BA.2.86 (including JN.1) variant from recent prior infection (OR = 2.8; 95% CI:1.2-6.5) and, not statistically significant, from XBB.1.5 vaccination (OR = 1.5; 95% CI:0.8-2.6).
Collapse
Affiliation(s)
- Anne J Huiberts
- Centre for Infectious Disease Control, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - Christina E Hoeve
- Centre for Infectious Disease Control, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - Brechje de Gier
- Centre for Infectious Disease Control, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - Jeroen Cremer
- Centre for Infectious Disease Control, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - Bas van der Veer
- Centre for Infectious Disease Control, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - Hester E de Melker
- Centre for Infectious Disease Control, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - Janneke Hhm van de Wijgert
- Centre for Infectious Disease Control, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht (UMCU), Utrecht, the Netherlands
| | - Susan van den Hof
- Centre for Infectious Disease Control, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - Dirk Eggink
- Centre for Infectious Disease Control, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
- These authors contributed equally to this article and share last authorship
| | - Mirjam J Knol
- Centre for Infectious Disease Control, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
- These authors contributed equally to this article and share last authorship
| |
Collapse
|
17
|
Cheng MQ, Li R, Weng ZY, Song G. Relative effectiveness of bivalent COVID-19 vaccine: a systematic review and meta-analysis. Front Med (Lausanne) 2024; 10:1322396. [PMID: 38384317 PMCID: PMC10879625 DOI: 10.3389/fmed.2023.1322396] [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: 10/16/2023] [Accepted: 12/14/2023] [Indexed: 02/23/2024] Open
Abstract
Objective The rapid development of COVID-19 bivalent vaccines (BVs) has encompassed both the original virus strains and the variant strain. However, the effectiveness of BVs is largely unknown. Therefore, we conducted a systematic review and meta-analysis of the effectiveness of BVs. Methods Literature research was conducted through PubMed, Cochrane Library, Embase, and Web of Science up until November 4, 2023. Both randomized control trials and observational studies were considered for inclusion. Pooled estimates were calculated using a random effects model. The Newcastle-Ottawa Scale (NOS) was used to assess the risk of bias in cohort and case-control studies. Results A total of 1,174 articles were reviewed and 22 eligible studies were included. All included studies were observational (15 cohort studies, 7 case-control studies). The total number of participants was 39,673,160, and the number of people vaccinated with BVs as an intervention group was 11,585,182. Two mRNA BVs were mainly involved, including the ancestral strain and the BA.1 or BA.4-5 variants. Meta-analysis results showed, compared with the monovalent vaccines (MVs), the relative effectiveness (rVE) of the BVs in COVID-19-associated infections/symptomatic infections, illnesses, hospitalizations, and deaths was 30.90% [95% confidence interval (CI), 8.43-53.37], 39.83% (95% CI, 27.34-52.32), 59.70% (95% CI, 44.08-75.32), and 72.23% (95% CI, 62.08-82.38), respectively. For those aged 50 years and older, BVs provided an additional 49.69% (95% CI, 41.44-57.94) effective protection compared with MVs. During the dominance period of the omicron XBB variant strain, BVs provided an additional 47.63% (95% CI, 27.45-67.82) effective protection compared with MVs. Conclusion Our findings show that the rVE of BVs in preventing COVID-19-associated infections, symptomatic infections, illnesses, hospitalizations, and deaths is higher compared to MVs. Particularly for people over 50 years of age and during the Omicron variant XBB dominance phase, BVs provided superior protection. Therefore, BVs may have a broader application in the prevention and control of coronaviruses variant.
Collapse
Affiliation(s)
- Meng-qun Cheng
- Department of Reproductive Medicine, The Puer People's Hospital, Pu’er, China
| | - Rong Li
- Department of Pharmacy, The Puer People's Hospital, Pu’er, China
| | - Zhi-ying Weng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Gao Song
- Department of Pharmacy, The Puer People's Hospital, Pu’er, China
| |
Collapse
|
18
|
Tamada Y, Takeuchi K, Kusama T, Maeda M, Murata F, Osaka K, Fukuda H. Bivalent mRNA vaccine effectiveness against COVID-19 among older adults in Japan: a test-negative study from the VENUS study. BMC Infect Dis 2024; 24:135. [PMID: 38287337 PMCID: PMC10823731 DOI: 10.1186/s12879-024-09035-3] [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: 11/18/2023] [Accepted: 01/19/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Bivalent COVID-19 vaccines have been implemented worldwide since the booster vaccination campaigns of autumn of 2022, but little is known about their effectiveness. Thus, this study holistically evaluated the effectiveness of bivalent vaccines against infection in older adults in Japan. METHODS We adopted the test-negative design using COVID-19 test data of individuals, aged ≥ 65 years, residing in three municipalities in Japan, who underwent tests in medical institutions between October 1 and December 30, 2022. Logistic regression analyses were conducted to estimate the odds of testing positive according to vaccination status. Vaccine effectiveness was defined as (1 - odds ratio) × 100%. RESULTS A total of 3,908 positive and 16,090 negative results were included in the analyses. Receiving a bivalent dose in addition to ≥ 2 monovalent doses was 33.6% (95% confidence interval [CI]: 20.8, 44.3%) more effective than receiving no vaccination, and 18.2% (95% CI: 9.4, 26.0%) more effective than receiving ≥ 2 monovalent doses but not receiving a bivalent vaccination. In addition, the effectiveness peaked at 14-20 days after administration and then gradually declined over time. Furthermore, a bivalent booster dose provided 18.6% (95% CI: 9.9, 26.5%) additional protection among those vaccinated with ≥ 2 monovalent doses, in the absence of a previous infection history. However, we did not find sufficient evidence of effectiveness of bivalent vaccines among previously infected older adults. CONCLUSIONS Bivalent vaccines are effective against COVID-19 infections among older adults without a history of infection.
Collapse
Affiliation(s)
- Yudai Tamada
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, 4-1, Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Kenji Takeuchi
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, 4-1, Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan.
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan.
- Division of Statistics and Data Science, Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Miyagi, Japan.
| | - Taro Kusama
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, 4-1, Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
- Division of Statistics and Data Science, Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Megumi Maeda
- Department of Health Care Administration and Management, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Fumiko Murata
- Department of Health Care Administration and Management, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Ken Osaka
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, 4-1, Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Haruhisa Fukuda
- Department of Health Care Administration and Management, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
19
|
Kirwan PD, Hall VJ, Foulkes S, Otter AD, Munro K, Sparkes D, Howells A, Platt N, Broad J, Crossman D, Norman C, Corrigan D, Jackson CH, Cole M, Brown CS, Atti A, Islam J, Presanis AM, Charlett A, De Angelis D, Hopkins S. Effect of second booster vaccinations and prior infection against SARS-CoV-2 in the UK SIREN healthcare worker cohort. THE LANCET REGIONAL HEALTH. EUROPE 2024; 36:100809. [PMID: 38111727 PMCID: PMC10727938 DOI: 10.1016/j.lanepe.2023.100809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/20/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023]
Abstract
Background The protection of fourth dose mRNA vaccination against SARS-CoV-2 is relevant to current global policy decisions regarding ongoing booster roll-out. We aimed to estimate the effect of fourth dose vaccination, prior infection, and duration of PCR positivity in a highly-vaccinated and largely prior-COVID-19 infected cohort of UK healthcare workers. Methods Participants underwent fortnightly PCR and regular antibody testing for SARS-CoV-2 and completed symptoms questionnaires. A multi-state model was used to estimate vaccine effectiveness (VE) against infection from a fourth dose compared to a waned third dose, with protection from prior infection and duration of PCR positivity jointly estimated. Findings 1298 infections were detected among 9560 individuals under active follow-up between September 2022 and March 2023. Compared to a waned third dose, fourth dose VE was 13.1% (95% CI 0.9 to 23.8) overall; 24.0% (95% CI 8.5 to 36.8) in the first 2 months post-vaccination, reducing to 10.3% (95% CI -11.4 to 27.8) and 1.7% (95% CI -17.0 to 17.4) at 2-4 and 4-6 months, respectively. Relative to an infection >2 years ago and controlling for vaccination, 63.6% (95% CI 46.9 to 75.0) and 29.1% (95% CI 3.8 to 43.1) greater protection against infection was estimated for an infection within the past 0-6, and 6-12 months, respectively. A fourth dose was associated with greater protection against asymptomatic infection than symptomatic infection, whilst prior infection independently provided more protection against symptomatic infection, particularly if the infection had occurred within the previous 6 months. Duration of PCR positivity was significantly lower for asymptomatic compared to symptomatic infection. Interpretation Despite rapid waning of protection, vaccine boosters remain an important tool in responding to the dynamic COVID-19 landscape; boosting population immunity in advance of periods of anticipated pressure, such as surging infection rates or emerging variants of concern. Funding UK Health Security Agency, Medical Research Council, NIHR HPRU Oxford, Bristol, and others.
Collapse
Affiliation(s)
- Peter D. Kirwan
- MRC Biostatistics Unit, University of Cambridge, United Kingdom
| | | | | | | | | | | | | | | | | | - David Crossman
- School of Medicine, University of St Andrews, United Kingdom
| | | | | | | | | | | | - Ana Atti
- UK Health Security Agency, United Kingdom
| | | | | | | | - Daniela De Angelis
- MRC Biostatistics Unit, University of Cambridge, United Kingdom
- UK Health Security Agency, United Kingdom
| | | |
Collapse
|
20
|
Meah S, Shi X, Fritsche LG, Salvatore M, Wagner A, Martin ET, Mukherjee B. Design and analysis heterogeneity in observational studies of COVID-19 booster effectiveness: A review and case study. SCIENCE ADVANCES 2023; 9:eadj3747. [PMID: 38117882 PMCID: PMC10732535 DOI: 10.1126/sciadv.adj3747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 11/16/2023] [Indexed: 12/22/2023]
Abstract
We investigated the design and analysis of observational booster vaccine effectiveness (VE) studies by performing a scoping review of booster VE literature with a focus on study design and analytic choices. We then applied 20 different approaches, including those found in the literature, to a single dataset from Michigan Medicine. We identified 80 studies in our review, including over 150 million observations in total. We found that while protection against infection is variable and dependent on several factors including the study population and time period, both monovalent boosters and particularly the bivalent booster offer strong protection against severe COVID-19. In addition, VE analyses with a severe disease outcome (hospitalization, intensive care unit admission, or death) appear to be more robust to design and analytic choices than an infection endpoint. In terms of design choices, we found that test-negative designs and their variants may offer advantages in statistical efficiency compared to cohort designs.
Collapse
Affiliation(s)
- Sabir Meah
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Xu Shi
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Lars G. Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Precision Health Data Science, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Maxwell Salvatore
- Center for Precision Health Data Science, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Abram Wagner
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Emily T. Martin
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Precision Health Data Science, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
21
|
Tan CY, Chiew CJ, Pang D, Lee VJ, Ong B, Wang LF, Ren EC, Lye DC, Tan KB. Effectiveness of bivalent mRNA vaccines against medically attended symptomatic SARS-CoV-2 infection and COVID-19-related hospital admission among SARS-CoV-2-naive and previously infected individuals: a retrospective cohort study. THE LANCET. INFECTIOUS DISEASES 2023; 23:1343-1348. [PMID: 37543042 DOI: 10.1016/s1473-3099(23)00373-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/22/2023] [Accepted: 06/08/2023] [Indexed: 08/07/2023]
Abstract
BACKGROUND Emergence of the SARS-CoV-2 omicron (B.1.1.529) variant with high immune evasion has led to the development and roll-out of bivalent mRNA vaccines targeting original and omicron strains. However, real-world observational data on effectiveness of bivalent vaccines are scarce. We aimed to assess the relative effectiveness of a fourth vaccine dose with the BA.1-adapted or BA.4/BA.5-adapted bivalent vaccines against medically attended symptomatic SARS-CoV-2 infection and COVID-19-related hospital admission among SARS-CoV-2-naive and previously infected individuals in Singapore. METHODS We conducted a retrospective cohort study among Singapore residents aged 18 years and older who had received three monovalent mRNA vaccine doses and were eligible for a fourth dose. Data were collected from official databases on COVID-19 cases and vaccinations maintained by the Singapore Ministry of Health. We analysed the incidence of medically attended symptomatic SARS-CoV-2 infection and COVID-19-related hospital admission between Oct 14, 2022, and Jan 31, 2023, by previous infection status and type of fourth vaccine dose received. Inverse probability-weighted Cox regressions were used to estimate hazard ratios (HRs). FINDINGS 2 749 819 individuals were included in the analysis. For the SARS-CoV-2-naive group, a fourth monovalent vaccine dose did not confer additional protection over three monovalent doses against symptomatic infection (HR 1·09 [95% CI 1·07-1·11]), whereas the bivalent vaccine did provide additional protection (0·18 [0·17-0·19]). Among individuals with previous infection, the HR was 0·87 (95% CI 0·84-0·91) and 0·14 (0·13-0·15) with receipt of the fourth monovalent and bivalent doses, respectively. Against COVID-19-related hospital admission, the bivalent vaccine (HR 0·12 [95% CI 0·08-0·18] in SARS-CoV-2-naive participants and 0·04 [0·01-0·15] in previously infected participants) conferred greater benefit compared with the fourth monovalent dose (0·84 [0·77-0·91] in SARS-CoV-2-naive participants and 0·85 [0·69-1·04] in previously infected participants). INTERPRETATION A fourth dose with the bivalent vaccine was substantially more effective against medically attended symptomatic SARS-CoV-2 infection and COVID-19-related hospital admission than four monovalent doses among both SARS-CoV-2-naive and previously infected individuals. Boosters with the bivalent vaccine might be preferred in this omicron-predominant pandemic, regardless of previous infection history. FUNDING None.
Collapse
Affiliation(s)
- Celine Y Tan
- Communicable Diseases Division, Ministry of Health, Singapore.
| | - Calvin J Chiew
- Communicable Diseases Division, Ministry of Health, Singapore; National Centre for Infectious Diseases, Singapore
| | - Deanette Pang
- Communicable Diseases Division, Ministry of Health, Singapore
| | - Vernon J Lee
- Communicable Diseases Division, Ministry of Health, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Benjamin Ong
- Communicable Diseases Division, Ministry of Health, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Ee Chee Ren
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Singapore Immunology Network, Agency for Science Technology and Research, Singapore
| | - David Chien Lye
- National Centre for Infectious Diseases, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore
| | - Kelvin Bryan Tan
- Communicable Diseases Division, Ministry of Health, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| |
Collapse
|
22
|
Chen SY, Lin CY, Chi H, Weng SL, Li ST, Tai YL, Huang YN, Huang H, Lin CH, Chiu NC. The Effectiveness of Bivalent COVID-19 Vaccination: A Preliminary Report. Life (Basel) 2023; 13:2094. [PMID: 37895475 PMCID: PMC10608313 DOI: 10.3390/life13102094] [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: 08/22/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Vaccination has been a game-changer in the long battle against COVID-19. However, waning vaccine-induced immunity and the immune evasion of emerging variants create challenges. The rapid-fire development of bivalent vaccines (BVs), comprising ancestral strains and a new variant, was authorized to prevent COVID-19, but the effectiveness of the updated vaccines remains largely unclear. Electronic databases were searched to investigate the immunogenicity and reactogenicity of BVs in humans. As of March 2023, 20 trials were identified. Compared with monovalent vaccination, the induced immunogenicity against ancestral strains was similar. The BVs demonstrated approximately 33-50% higher immunogenicity values against additional variant strains. An observational cohort study showed the additional clinical effectiveness of the BVs. The adverse events were similar. In conclusion, our systematic review found that the BVs had equal immunogenicity against ancestral strains without safety concerns. Approximately 33-50% increased additional antibody titers and clinical effectiveness against additional variant strains were observed in subjects with a BV vaccine with moderate heterogeneity, especially for BA.1-containing BVs.
Collapse
Affiliation(s)
- Ssu-Yu Chen
- Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan
- Hsinchu Municipal MacKay Children’s Hospital, Hsinchu City 300, Taiwan
| | - Chien-Yu Lin
- Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan
- Hsinchu Municipal MacKay Children’s Hospital, Hsinchu City 300, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City 251, Taiwan
| | - Hsin Chi
- Department of Medicine, MacKay Medical College, New Taipei City 251, Taiwan
- MacKay Children’s Hospital, Taipei 104, Taiwan
| | - Shun-Long Weng
- Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan
- Hsinchu Municipal MacKay Children’s Hospital, Hsinchu City 300, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City 251, Taiwan
| | - Sung-Tse Li
- Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan
- Hsinchu Municipal MacKay Children’s Hospital, Hsinchu City 300, Taiwan
| | - Yu-Lin Tai
- Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan
- Hsinchu Municipal MacKay Children’s Hospital, Hsinchu City 300, Taiwan
| | - Ya-Ning Huang
- Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan
- Hsinchu Municipal MacKay Children’s Hospital, Hsinchu City 300, Taiwan
| | - Hsiang Huang
- Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan
- Hsinchu Municipal MacKay Children’s Hospital, Hsinchu City 300, Taiwan
| | - Chao-Hsu Lin
- Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan
- Hsinchu Municipal MacKay Children’s Hospital, Hsinchu City 300, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City 251, Taiwan
| | - Nan-Chang Chiu
- Department of Medicine, MacKay Medical College, New Taipei City 251, Taiwan
- MacKay Children’s Hospital, Taipei 104, Taiwan
| |
Collapse
|
23
|
Matsumoto N, Mitsuhashi T, Matsuo R, Kadowaki T, Takao S, Yorifuji T. Effectiveness of the Original COVID-19 Vaccine against COVID-19 Exacerbations during the Omicron Wave: A Population-based Study in Okayama, Japan. JMA J 2023; 6:463-469. [PMID: 37941689 PMCID: PMC10628327 DOI: 10.31662/jmaj.2023-0019] [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: 02/10/2023] [Accepted: 08/07/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction In Japan, approximately 97 million individuals have received their primary two doses of coronavirus disease 2019 (COVID-19) vaccine at the end of 2022. In this study, we aim to examine the effectiveness of the primary vaccines and compare its efficacy to booster vaccine shots in terms of preventing COVID-19 exacerbations during the Omicron-predominant period in Japan. Methods For this analysis, we have collected all the confirmed COVID-19-positive cases from different medical institutions in Okayama City and have also utilized the information from the public Vaccination Record System. Taking the number of vaccinations into consideration, we then conducted a population-based study to assess the effectiveness of the two primary vaccine doses in preventing COVID-19 exacerbations during the Omicron waves. Our primary and secondary outcomes were COVID-19 exacerbations with respiratory failure (i.e., oxygen saturation on room air ≤ 93%, requiring supplemental oxygen), intensive care unit admission and/or mechanical ventilator requirement, or death, in accordance with the Japanese COVID-19 guidelines, and pneumonia during the course of COVID-19 infection, respectively. Results In total, 95,329 COVID-19-positive individuals, aged 5 years and above, were included in this analysis (study period from January 1 to September 10, 2022). As per our findings, the effectiveness of the primary two doses against COVID-19 exacerbations compared with those who had never been vaccinated was 55.5% (95% confidential interval [CI]: 32.6-71.7), whereas it was higher after the third dose (76.9%; 95% CI: 66.7-84.0) and the fourth dose (75.7%; 95% CI: 58.8-85.7). Effectiveness was sustained for ≥ 5 months after the third vaccination, and preventive effectiveness was observed in individuals aged ≥ 65 years. Conclusions As per the results of this study, we can conclude that the efficacy of the primary two doses of SARS-CoV-2 vaccine can be further strengthened in terms of preventing COVID-19 exacerbations by administering third and fourth booster vaccine shots. The additional bivalent vaccine is anticipated to further increase its efficacy against the Omicron strain, suggesting that individuals who have not received their booster shots yet should consider getting them to prevent COVID-19 exacerbations.
Collapse
Affiliation(s)
- Naomi Matsumoto
- Department of Epidemiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toshiharu Mitsuhashi
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Rumi Matsuo
- Department of Epidemiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tomoka Kadowaki
- Department of Epidemiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Soshi Takao
- Department of Epidemiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takashi Yorifuji
- Department of Epidemiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
24
|
Meeraus W, Stuurman AL, Durukal I, Conde-Sousa E, Lee A, Maria AS, Furtado BE, Ouwens M, Gray CM, Valverde DA, da Silva HG, Taylor S. COVID-19 vaccine booster doses provide increased protection against COVID-19 hospitalization compared with previously vaccinated individuals: Interim findings from the REFORCO-Brazil real-world effectiveness study during Delta and Omicron. Vaccine 2023; 41:6366-6378. [PMID: 37704499 DOI: 10.1016/j.vaccine.2023.08.085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/15/2023] [Accepted: 08/30/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Although COVID-19 booster vaccination is widely recommended, there is limited long-term, population-level, real-world evidence on the magnitude of improved protection against severe COVID-19 conferred by boosting with monovalent COVID-19 vaccines developed against ancestral SARS-CoV-2, especially in low- or middle-income countries. We present interim results from the first large-scale assessment of the relative vaccine effectiveness (rVE) of first and second booster doses against severe COVID-19 in a low-/middle-income country. METHODS REFORCO-Brazil is an ongoing, test-negative case-control study (NCT05697705) utilizing Brazil national severe acute respiratory syndrome (SARS) surveillance and vaccination data. In SARS hospitalizations from August 1, 2021 to July 31, 2022, we matched test-positive (via SARS-CoV-2 antigen/reverse transcription polymerase chain reaction [RT-PCR]) cases and test-negative case-controls (via RT-PCR) based on admission date, preceding vaccinations, and age. We evaluated the rVEs of four monovalent COVID-19 vaccines (AZD1222, Ad26.COV2.S, CoronaVac, and BNT162b2) as second boosters compared with any first boosters received ≥4 months previously, and as first boosters compared with primary-series vaccinations completed ≥4 months previously. RESULTS The overall rVE of second boosters, from 5668 (2238 test-positive) evaluated hospitalizations, was 24.7 % (95 % confidence interval [CI]: 12.6-35.1); the overall rVE of first boosters, from 30,272 (12,063 test-positive) hospitalizations, was 46.8 % (95 % CI: 43.3-50.0). The rVEs of AZD1222 and BNT162b2 were similar: 29.4 % (95 % CI: 8.6-45.5) and 25.5 % (95 % CI: 4.2-42.2), respectively, for second boosters; and 42.5 % (95 % CI: 28.0-54.0) and 50.8 % (95 % CI: 47.5-54.0), respectively, for first boosters. In general, rVEs were higher in elderly (≥80 years) and immunocompromised/high-risk individuals. CONCLUSIONS Our results support the use of AZD1222 and other adenoviral/mRNA vaccine boosters to maintain protection against COVID-19 hospitalization from Omicron subvariants, including in elderly and immunocompromised individuals at increased risk of accelerated waning or severe outcomes.
Collapse
Affiliation(s)
- Wilhelmine Meeraus
- Medical Evidence, Vaccines & Immune Therapies, AstraZeneca, Cambridge, UK.
| | - Anke L Stuurman
- Medical Evidence, Vaccines & Immune Therapies, AstraZeneca, Cambridge, UK; P95 Epidemiology & Pharmacovigilance, Leuven, Belgium
| | - Ilgaz Durukal
- Real World Science, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | - Eduardo Conde-Sousa
- Medical Evidence, Vaccines & Immune Therapies, AstraZeneca, Cambridge, UK; P95 Epidemiology & Pharmacovigilance, Leuven, Belgium
| | - Andrew Lee
- Medical and Payor Statistics, BioPharmaceuticals Business Unit, AstraZeneca, Cambridge, UK
| | | | | | - Mario Ouwens
- Medical and Payor Statistics, BioPharmaceuticals Business Unit, AstraZeneca, Mölndal, Sweden
| | - Christen M Gray
- Real World Science, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | | | | | - Sylvia Taylor
- Medical Evidence, Vaccines & Immune Therapies, AstraZeneca, Cambridge, UK
| |
Collapse
|
25
|
Chemaitelly H, Ayoub HH, AlMukdad S, Faust JS, Tang P, Coyle P, Yassine HM, Al Thani AA, Al-Khatib HA, Hasan MR, Al-Kanaani Z, Al-Kuwari E, Jeremijenko A, Kaleeckal AH, Latif AN, Shaik RM, Abdul-Rahim HF, Nasrallah GK, Al-Kuwari MG, Butt AA, Al-Romaihi HE, Al-Thani MH, Al-Khal A, Bertollini R, Abu-Raddad LJ. Bivalent mRNA-1273.214 vaccine effectiveness against SARS-CoV-2 omicron XBB* infections. J Travel Med 2023; 30:taad106. [PMID: 37555656 PMCID: PMC10481416 DOI: 10.1093/jtm/taad106] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/06/2023] [Accepted: 08/07/2023] [Indexed: 08/10/2023]
Abstract
Effectiveness of the 50-μg mRNA-1273.214 bivalent vaccine against SARS-CoV-2 infection was modest at 25% in a matched, retrospective, cohort study in Qatar comparing infection incidence in the bivalent cohort to that in the national no-recent-vaccination resident cohort. XBB* immune evasion, immune imprinting effects, or both, may explain findings.
Collapse
Affiliation(s)
- Hiam Chemaitelly
- Infectious Disease Epidemiology Group, Weill Cornell Medicine-Qatar, Cornell University, PO Box 24144, Doha, Qatar
| | - Houssein H Ayoub
- Departments of Mathematics, Statistics, and Physics, and of Biomedical Science, and of Public Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Sawsan AlMukdad
- Infectious Disease Epidemiology Group, Weill Cornell Medicine-Qatar, Cornell University, PO Box 24144, Doha, Qatar
| | - Jeremy S Faust
- Department of Emergency Medicine, Brigham and Women’s Hospital, MA 02115, Boston, Massachusetts, USA
| | - Patrick Tang
- Department of Pathology, Sidra Medicine, PO Box 26999, Doha
| | - Peter Coyle
- Hamad Medical Corporation, POBox 3050, Doha, Qatar
| | - Hadi M Yassine
- Departments of Mathematics, Statistics, and Physics, and of Biomedical Science, and of Public Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Asmaa A Al Thani
- Departments of Mathematics, Statistics, and Physics, and of Biomedical Science, and of Public Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Hebah A Al-Khatib
- Departments of Mathematics, Statistics, and Physics, and of Biomedical Science, and of Public Health, Qatar University, PO Box 2713, Doha, Qatar
| | | | | | | | | | | | - Ali N Latif
- Hamad Medical Corporation, POBox 3050, Doha, Qatar
| | | | - Hanan F Abdul-Rahim
- Departments of Mathematics, Statistics, and Physics, and of Biomedical Science, and of Public Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Gheyath K Nasrallah
- Departments of Mathematics, Statistics, and Physics, and of Biomedical Science, and of Public Health, Qatar University, PO Box 2713, Doha, Qatar
| | | | - Adeel A Butt
- Hamad Medical Corporation, POBox 3050, Doha, Qatar
| | | | | | | | | | - Laith J Abu-Raddad
- Infectious Disease Epidemiology Group, Weill Cornell Medicine-Qatar, Cornell University, PO Box 24144, Doha, Qatar
| |
Collapse
|
26
|
Lippi G, Henry BM. Understanding the biological success of SARS-CoV-2: Immunoevasion strategies and beyond. Eur J Intern Med 2023; 114:37-39. [PMID: 37277247 PMCID: PMC10235673 DOI: 10.1016/j.ejim.2023.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/07/2023]
Affiliation(s)
- Giuseppe Lippi
- Section of Clinical Biochemistry and School of Medicine, University of Verona, Verona, Italy.
| | - Brandon M Henry
- Clinical Laboratory, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| |
Collapse
|
27
|
Andersson NW, Thiesson EM, Baum U, Pihlström N, Starrfelt J, Faksová K, Poukka E, Meijerink H, Ljung R, Hviid A. Comparative effectiveness of bivalent BA.4-5 and BA.1 mRNA booster vaccines among adults aged ≥50 years in Nordic countries: nationwide cohort study. BMJ 2023; 382:e075286. [PMID: 37491022 PMCID: PMC10364194 DOI: 10.1136/bmj-2022-075286] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
OBJECTIVE To estimate the effectiveness of the bivalent mRNA booster vaccines containing the original SARS-CoV-2 and omicron BA.4-5 or BA.1 subvariants as the fourth dose against severe covid-19. DESIGN Nationwide cohort analyses, using target trial emulation. SETTING Denmark, Finland, Norway, and Sweden, from 1 July 2022 to 10 April 2023. PARTICIPANTS People aged ≥50 years who had received at least three doses of covid-19 vaccine (that is, a primary course and a first booster). MAIN OUTCOME MEASURES The Kaplan-Meier estimator was used to compare the risk of hospital admission and death related to covid-19 in people who received a bivalent Comirnaty (Pfizer-BioNTech) or Spikevax (Moderna) BA.4-5 or BA.1 mRNA booster vaccine as a fourth dose (second booster) with three dose (first booster) vaccinated people and between four dose vaccinated people. RESULTS A total of 1 634 199 people receiving bivalent BA.4-5 fourth dose booster and 1 042 124 receiving bivalent BA.1 fourth dose booster across the four Nordic countries were included. Receipt of a bivalent BA.4-5 booster as a fourth dose was associated with a comparative vaccine effectiveness against admission to hospital with covid-19 of 67.8% (95% confidence interval 63.1% to 72.5%) and a risk difference of -91.9 (95% confidence interval -152.4 to -31.4) per 100 000 people at three months of follow-up compared with having received three doses of vaccine (289 v 893 events). The corresponding comparative vaccine effectiveness and risk difference for bivalent BA.1 boosters (332 v 977 events) were 65.8% (59.1% to 72.4%) and -112.9 (-179.6 to -46.2) per 100 000, respectively. Comparative vaccine effectiveness and risk difference against covid-19 related death were 69.8% (52.8% to 86.8%) and -34.1 (-40.1 to -28.2) per 100 000 for bivalent BA.4-5 booster (93 v 325 events) and 70.0% (50.3% to 89.7%) and -38.7 (-65.4 to -12.0) per 100 000 for BA.1 booster (86 v 286) as a fourth dose. Comparing bivalent BA.4-5 and BA.1 boosters as a fourth dose directly resulted in a three month comparative vaccine effectiveness and corresponding risk difference of -14.9% (-62.3% to 32.4%) and 10.0 (-14.4 to 34.4) per 100 000 people for admission to hospital with covid-19 (802 v 932 unweighted events) and -40.7% (-123.4% to 42.1%) and 8.1 (-3.3 to 19.4) per 100 000 for covid-19 related death (229 v 243 unweighted events). The comparative vaccine effectiveness did not differ across sex and age (</≥70 years) and seemed to be sustained up to six months from the day of vaccination with modest waning. CONCLUSION Vaccination with bivalent BA.4-5 or BA.1 mRNA booster vaccines as a fourth dose was associated with reduced rates of covid-19 related hospital admission and death among adults aged ≥50 years. The protection afforded by the bivalent BA.4-5 and BA.1 boosters did not differ significantly when directly compared, and any potential difference would most likely be very small in absolute numbers.
Collapse
Affiliation(s)
| | | | - Ulrike Baum
- Infectious Disease Control and Vaccinations Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Nicklas Pihlström
- Division of Licensing, Swedish Medical Products Agency, Uppsala, Sweden
| | - Jostein Starrfelt
- Department of Infection Control and Preparedness, Norwegian Institute of Public Health, Oslo, Norway
| | - Kristýna Faksová
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Eero Poukka
- Infectious Disease Control and Vaccinations Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Hinta Meijerink
- Department of Infection Control and Vaccines, Norwegian Institute of Public Health, Oslo, Norway
| | - Rickard Ljung
- Division of Use and Information, Swedish Medical Products Agency, Uppsala, Sweden
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anders Hviid
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Pharmacovigilance Research Center, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Meah S, Shi X, Fritsche LG, Salvatore M, Wagner A, Martin ET, Mukherjee B. Design and Analysis Heterogeneity in Observational Studies of COVID-19 Booster Effectiveness: A Review and Case Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.22.23291692. [PMID: 37425863 PMCID: PMC10327238 DOI: 10.1101/2023.06.22.23291692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Background Observational vaccine effectiveness (VE) studies based on real-world data are a crucial supplement to initial randomized clinical trials of Coronavirus Disease 2019 (COVID-19) vaccines. However, there exists substantial heterogeneity in study designs and statistical methods for estimating VE. The impact of such heterogeneity on VE estimates is not clear. Methods We conducted a two-step literature review of booster VE: a literature search for first or second monovalent boosters on January 1, 2023, and a rapid search for bivalent boosters on March 28, 2023. For each study identified, study design, methods, and VE estimates for infection, hospitalization, and/or death were extracted and summarized via forest plots. We then applied methods identified in the literature to a single dataset from Michigan Medicine (MM), providing a comparison of the impact of different statistical methodologies on the same dataset. Results We identified 53 studies estimating VE of the first booster, 16 for the second booster. Of these studies, 2 were case-control, 17 were test-negative, and 50 were cohort studies. Together, they included nearly 130 million people worldwide. VE for all outcomes was very high (around 90%) in earlier studies (i.e., in 2021), but became attenuated and more heterogeneous over time (around 40%-50% for infection, 60%-90% for hospitalization, and 50%-90% for death). VE compared to the previous dose was lower for the second booster (10-30% for infection, 30-60% against hospitalization, and 50-90% against death). We also identified 11 bivalent booster studies including over 20 million people. Early studies of the bivalent booster showed increased effectiveness compared to the monovalent booster (VE around 50-80% for hospitalization and death).Our primary analysis with MM data using a cohort design included 186,495 individuals overall (including 153,811 boosted and 32,684 with only a primary series vaccination), and a secondary test-negative design included 65,992 individuals tested for SARS-CoV-2. When different statistical designs and methods were applied to MM data, VE estimates for hospitalization and death were robust to analytic choices, with test-negative designs leading to narrower confidence intervals. Adjusting either for the propensity of getting boosted or directly adjusting for covariates reduced the heterogeneity across VE estimates for the infection outcome. Conclusion While the advantage of the second monovalent booster is not obvious from the literature review, the first monovalent booster and the bivalent booster appear to offer strong protection against severe COVID-19. Based on both the literature view and data analysis, VE analyses with a severe disease outcome (hospitalization, ICU admission, or death) appear to be more robust to design and analytic choices than an infection endpoint. Test-negative designs can extend to severe disease outcomes and may offer advantages in statistical efficiency when used properly.
Collapse
Affiliation(s)
- Sabir Meah
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Xu Shi
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Lars G. Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Precision Health Data Science, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Maxwell Salvatore
- Center for Precision Health Data Science, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Abram Wagner
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Emily T. Martin
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Precision Health Data Science, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| |
Collapse
|
29
|
Hirotsu Y, Sugiura H, Takatori M, Mochizuki H, Omata M. Antibody Response to the BA.5 Bivalent Vaccine Shot: a Two-Year Follow-Up Study following Initial COVID-19 mRNA Vaccination. Microbiol Spectr 2023; 11:e0131623. [PMID: 37191496 PMCID: PMC10269437 DOI: 10.1128/spectrum.01316-23] [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: 03/27/2023] [Accepted: 04/27/2023] [Indexed: 05/17/2023] Open
Abstract
Although many studies have been conducted on the increase in spike antibody levels after vaccination, there is insufficient prospective and longitudinal information on the BA.5-adapted bivalent vaccine up to the fifth vaccination. In this study, we conducted a follow-up study of spike antibody levels and infection history in 46 health care workers who received up to 5 vaccinations. Monovalent vaccines were administered for the first to fourth vaccinations, and a bivalent vaccine was administered for the fifth vaccination. 11 serum samples were collected from each participant, and antibody levels were measured in a total of 506 serum samples. During the observation period, 43 of the 46 health care workers had no infection history, and 3 had a history of infection. Spike antibody levels peaked at 1 week after the second booster vaccination and gradually declined until the 27th week after the second vaccination. After 2 weeks following the fifth BA.5-adapted bivalent vaccine, the spike antibody levels significantly increased (median: 23,756 [IQR: 16,450 to 37,326]), compared to those measured before vaccination (median: 9,354 [IQR: 5,904 to 15,784]) (paired Wilcoxon signed-rank test, P = 5.7 × 10-14). These changes in antibody kinetics were observed regardless of age or sex. These results suggest that booster vaccination increased the spike antibody levels. Regular vaccination is effective in maintaining long-term antibody levels. IMPORTANCE A COVID-19 bivalent mRNA vaccine was developed and administered to health care workers. The COVID-19 mRNA vaccine induces a robust antibody response. However, little is known about the antibody response to vaccines in serially collected blood samples from the same individuals. Here, we provide two-year follow-up data on the humoral immune response to COVID-19 mRNA vaccines in health care workers who received up to five vaccinations, including the BA.5-adapted bivalent vaccine. The results suggest that regular vaccination is effective in maintaining long-term antibody levels and have implications for vaccine efficacy and booster dose strategies in health care settings.
Collapse
Affiliation(s)
- Yosuke Hirotsu
- Genome Analysis Center, Yamanashi Central Hospital, Kofu, Japan
| | - Hiroki Sugiura
- Division of Clinical Biochemistry and Immunology, Yamanashi Central Hospital, Kofu, Japan
| | - Mika Takatori
- Division of Infection Control and Prevention, Yamanashi Central Hospital, Kofu, Japan
| | - Hitoshi Mochizuki
- Genome Analysis Center, Yamanashi Central Hospital, Kofu, Japan
- Central Clinical Laboratory, Yamanashi Central Hospital, Kofu, Japan
- Department of Gastroenterology, Yamanashi Central Hospital, Kofu, Japan
| | - Masao Omata
- Department of Gastroenterology, Yamanashi Central Hospital, Kofu, Japan
- The University of Tokyo, Tokyo, Japan
| |
Collapse
|