1
|
Hwang YH, Min DH, Beom Park W. Limitations of neutralizing antibody titers in COVID-19 vaccine efficacy trials and a call for additional correlates of protection. Hum Vaccin Immunother 2025; 21:2473795. [PMID: 40051347 PMCID: PMC11901426 DOI: 10.1080/21645515.2025.2473795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The coronavirus disease (COVID-19) pandemic accelerated development of various vaccine platforms. Among them, mRNA vaccines played a crucial role in controlling the pandemic due to their swift development and efficacy against virus variants. Despite the success of these vaccines, recent studies highlight challenges in evaluating vaccine efficacy, especially in individuals with prior COVID-19 infection. Weakened neutralizing antibody responses after additional doses are observed in these populations, raising concerns about using neutralizing antibody titers as the sole immune correlate of protection. While neutralizing antibodies remain the primary endpoint in immunogenicity trials, they may not fully capture the immune response in populations with widespread prior infection or vaccination. This review explores reduced neutralizing antibody responses in previously infected individuals, and their impact on vaccine efficacy evaluation. It also offers recommendations for improving efficacy assessment, stressing incorporation of additional immune markers such as cell-mediated immunity to enable more comprehensive understanding of vaccine-induced immunity.
Collapse
Affiliation(s)
- Young Hoon Hwang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dal-Hee Min
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Wan Beom Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Zanella MC, Rhee C, Klompas M. Preventing respiratory viral transmission in healthcare settings. Curr Opin Infect Dis 2025:00001432-990000000-00228. [PMID: 40314314 DOI: 10.1097/qco.0000000000001115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
PURPOSE OF REVIEW The COVID-19 pandemic catalyzed new insights into respiratory viral transmission mechanisms and prevention. We review the most practical and impactful measures to prevent SARS-CoV-2 and other nosocomial respiratory viral infections in healthcare. RECENT FINDINGS Nosocomial respiratory viral infection rates mirror viral activity levels in the surrounding community. During peak periods ∼15-20% of hospitalized patients with respiratory viral infections may have acquired their virus in the hospital. Nosocomial respiratory viral infections are associated with increased lengths-of-stay, risk of respiratory failure, and hospital death. Most procedures contribute minimally to aerosol production compared to labored breathing, coughing, and forced expiration. Masking for source control and exposure control both decrease transmission risk, respirators more so than masks. Likewise, vaccinating healthcare workers decreases transmission risk and is associated with lower patient mortality rates, particularly in long-term care facilities. Increasing air changes, ultraviolet irradiation, and portable HEPA filtration units may also decrease transmission rates but their marginal benefit relative to current healthcare ventilation standards has yet to be established. SUMMARY Practical strategies to prevent nosocomial respiratory viral infections include maximizing staff and patient influenza and SARS-CoV-2 vaccination rates and implementing routine masking during patient interactions when community incidence is high.
Collapse
Affiliation(s)
- Marie-Céline Zanella
- Infection Control Program and WHO Collaborating Centre, Faculty of Medicine, University of Geneva Hospitals, Geneva, Switzerland
| | - Chanu Rhee
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Healthcare Institute
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Michael Klompas
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Healthcare Institute
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Okoro EO, Ikoba NA, Okoro BE, Akpila AS, Salihu MO. Paradoxical increase in global COVID-19 deaths with vaccination coverage: World Health Organization estimates (2020-2023). INTERNATIONAL JOURNAL OF RISK & SAFETY IN MEDICINE 2025:9246479251336610. [PMID: 40265700 DOI: 10.1177/09246479251336610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
BackgroundMany reports on the impact of vaccination on COVID-19 pandemic deaths were projections undertaken as the global emergency was unfolding. An increasing number of independent investigators have drawn attention to the subjective nature and inherent biases in mathematical models used for such forecasts that could undermine their accuracy when excess mortality was the metric of choice.ObjectiveCOVID-19 deaths were compared between the pre-vaccines and vaccination eras to observe how vaccination impacted COVID-19 death trajectory worldwide during the pandemic emergency.MethodsCOVID-19 cases, deaths and vaccination rates in World Health Organization (WHO) database till 07 June 2023, Case fatality rate per 1000 for the pre-vaccines period (CFR1), and that over vaccination era (CFR2) were compared for all WHO regions, while tests of correlation between the percentage change in COVID-19 deaths and variables of interest were examined.ResultsCOVID-19 deaths increased with vaccination coverage ranging from 43.3% (Africa) to 1275.0% (Western Pacific). The Western Pacific (1.5%) and Africa (3.8%) regions contributed least to the global cumulative COVID-19 deaths pre-vaccines, while the Americas (49.9%) and Europe (27.6%) had the highest counts. The Americas (39.8%) and Europe (34.1%) accounted for >70% of global COVID-19 deaths despite high vaccination, and the percentage increase in COVID-19 mortality and the percentage of person's ≥65 years were significantly correlated (0.48) in Africa.ConclusionCOVID-19 mortality increased in the vaccination era, especially in regions with higher vaccination coverage.
Collapse
Affiliation(s)
- Emmanuel O Okoro
- Department of Medicine, University of Ilorin Teaching Hospital, Ilorin, Nigeria
- Department of Medicine, University of Ilorin, Ilorin, Nigeria
| | | | | | - Azibanigha S Akpila
- Department of Medicine, University of Ilorin Teaching Hospital, Ilorin, Nigeria
- Department of Obstetrics and Gynecology, Mersey and West Lancashire Teaching Hospital, NHS Trust, Wirral, UK
| | - Mumeen O Salihu
- Department of Behavioral Sciences, University of Ilorin Teaching Hospital, Ilorin, Nigeria
- Department of Behavioral Sciences, Kwara State University Teaching Hospital, Ilorin, Nigeria
| |
Collapse
|
4
|
Molinos-Albert LM, Rubio R, Martín-Pérez C, Pradenas E, Torres C, Jiménez A, Canyelles M, Vidal M, Barrios D, Marfil S, Aparicio E, Ramírez-Morros A, Trinité B, Vidal-Alaball J, Santamaria P, Serra P, Izquierdo L, Aguilar R, Ruiz-Comellas A, Blanco J, Dobaño C, Moncunill G. Long-lasting antibody B-cell responses to SARS-CoV-2 three years after the onset of the pandemic. Cell Rep 2025; 44:115498. [PMID: 40173043 DOI: 10.1016/j.celrep.2025.115498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/21/2024] [Accepted: 03/11/2025] [Indexed: 04/04/2025] Open
Abstract
Immune memory is essential for the effectiveness of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination. In the current context of the pandemic, with a diminished vaccine efficacy against emerging variants, it remains crucial to perform long-term studies to evaluate the durability and quality of immune responses. Here, we examined the antibody and memory B-cell responses in a cohort of 113 healthcare workers with distinct exposure histories over a 3-year period. Previously infected and naive participants developed comparable humoral responses by 17 months after receiving a full three-dose mRNA vaccination. In addition, both maintained a substantial SARS-CoV-2-reactive memory B-cell pool, associated with a lower incidence of breakthrough infections in naive participants. Of note, previously infected participants developed an expanded SARS-CoV-2-reactive CD27-CD21- atypical B-cell population that remained stable throughout the follow-up period. Thus, previous SARS-CoV-2 infection differentially imprints the memory B-cell compartment without compromising the development of long-lasting humoral responses.
Collapse
Affiliation(s)
- Luis M Molinos-Albert
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.
| | - Rocío Rubio
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Carla Martín-Pérez
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Edwards Pradenas
- IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona (Barcelona), Spain
| | - Cèlia Torres
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Alfons Jiménez
- ISGlobal, Barcelona, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Mar Canyelles
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Marta Vidal
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | | | - Silvia Marfil
- IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona (Barcelona), Spain
| | - Ester Aparicio
- IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona (Barcelona), Spain
| | - Anna Ramírez-Morros
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la Recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJG), Manresa, Spain
| | - Benjamin Trinité
- IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona (Barcelona), Spain
| | - Josep Vidal-Alaball
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la Recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJG), Manresa, Spain; Health Promotion in Rural Areas Research Group (PROSAARU), Gerència Territorial de la Catalunya Central, Institut Català de la Salut, Manresa, Spain; Universitat de Vic-Universitat Central de Catalunya (uVic-UCC), Vic, Spain
| | - Pere Santamaria
- Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Department of Microbiology, Immunology, and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Pau Serra
- Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Luis Izquierdo
- ISGlobal, Barcelona, Spain; CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Ruth Aguilar
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Anna Ruiz-Comellas
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la Recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJG), Manresa, Spain; Health Promotion in Rural Areas Research Group (PROSAARU), Gerència Territorial de la Catalunya Central, Institut Català de la Salut, Manresa, Spain; Centre d'Atenció Primària (CAP) Sant Joan de Vilatorrada, Gerència Territorial de la Catalunya Central, Institut Català de la Salut (ICS), Manresa, Spain; Universitat de Vic-Universitat Central de Catalunya (uVic-UCC), Vic, Spain
| | - Julià Blanco
- IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona (Barcelona), Spain; CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; Universitat de Vic-Universitat Central de Catalunya (uVic-UCC), Vic, Spain; Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, Badalona (Barcelona), Spain
| | - Carlota Dobaño
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain; CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| | - Gemma Moncunill
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain; CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
5
|
Chitre S, Barrie MB, Kanu JS, Conteh TS, Bayoh M, Kamara MN, Bangura HF, Lascher JS, Frankfurter R, Goldberg SA, Glidden DV, Kelly JD, Lakoh S, Richardson ET. Post-Omicron SARS-CoV-2 antibody prevalence in Sierra Leone: A cross-sectional, nationally representative, follow-up serosurvey. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0004273. [PMID: 40238730 PMCID: PMC12002446 DOI: 10.1371/journal.pgph.0004273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/22/2025] [Indexed: 04/18/2025]
Abstract
Based on a serosurvey conducted in March 2021, Barrie and colleagues published the first nationally representative SARS-CoV-2 serosurvey in Africa, estimating a SARS-CoV-2 seroprevalence of 2.6% in Sierra Leone, 43 times higher than the reported number of cases at that time. Over the following two years, increasingly transmissible variants-specifically Delta and Omicron-proliferated across the globe, and their impact in Africa is poorly understood. Additional nationally representative seroprevalence data are therefore necessary to understand the pandemic's progression on the continent and for evaluating containment measures and future preparedness. Our follow-up nationally representative survey was conducted in Sierra Leone from February to March 2023. We returned to the 120 Enumeration Areas throughout the country collecting blood samples from one or more individuals per household as well as information on sociodemographic characteristics, history of COVID-19 infection and immunization, and attitudes towards vaccination. The weighted overall seroprevalence (vaccinated and/or SARS-CoV-2 infection) for individuals >19 years of age was 33% (95% CI 29-37). Using the data and distributions from our previous serosurvey, the weighted predicted seroprevalence (any prior SARS-CoV-2 infection) for the general population was 28% (95% CI 15-41). The weighted predicted seroprevalence was ~11 times higher than the pre-Delta/Omicron prevalence. It was also over 300 times higher than the reported number of cases. Despite this, overall seroprevalence was low compared with countries in Europe and the Americas (pointing towards lower transmission in Sierra Leone). In addition, our results suggest the following regarding prevention campaigns claiming to have vaccinated 70% of adults in Sierra Leone as of December 2022: 1) they resulted in limited seroconversion; 2) there was significant waning of immunity; and/or 3) many less individuals were vaccinated than reported. Regardless of the cause, the utility of COVID-19 Vaccine Delivery Partnership (CoVDP) efforts three years into the pandemic is called into question.
Collapse
Affiliation(s)
- Smit Chitre
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mohamed Bailor Barrie
- Partners In Health, Kono, Sierra Leone
- Institute for Global Health Sciences, University of California, San Francisco, California, United States of America
| | - Joseph Sam Kanu
- Ministry of Health and Sanitation, Government of Sierra Leone, Freetown, Sierra Leone
- College of Medicine and Allied Health Sciences, University of Sierra Leone, Freetown, Sierra Leone
| | - Theophilus S. Conteh
- Ministry of Health and Sanitation, Government of Sierra Leone, Freetown, Sierra Leone
| | | | - Matilda N. Kamara
- Ministry of Health and Sanitation, Government of Sierra Leone, Freetown, Sierra Leone
| | - Haja Fatmata Bangura
- Ministry of Health and Sanitation, Government of Sierra Leone, Freetown, Sierra Leone
| | - Jonathan S. Lascher
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Raphael Frankfurter
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Sarah A. Goldberg
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
| | - David V. Glidden
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
| | - J. Daniel Kelly
- Institute for Global Health Sciences, University of California, San Francisco, California, United States of America
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
| | - Sulaiman Lakoh
- Ministry of Health and Sanitation, Government of Sierra Leone, Freetown, Sierra Leone
- College of Medicine and Allied Health Sciences, University of Sierra Leone, Freetown, Sierra Leone
| | - Eugene T. Richardson
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| |
Collapse
|
6
|
Rudan I, Kerr S, Simpson CR, Kurdi A, Adeloye D, Robertson C, Sheikh A, EAVE II Collaboration. The COVID-19 pandemic in children and young people during 2022-24: what new did we learn? J Glob Health 2025; 15:01002. [PMID: 40168509 PMCID: PMC11961057 DOI: 10.7189/jogh.15.01002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025] Open
Abstract
The research conducted between 2022 and 2024 has advanced our understanding of COVID-19 in children and young people (CYP), particularly with the emergence of the Omicron variant and its subvariants. The findings have reinforced that, while Omicron infections are often milder compared to earlier variants, the overall seroprevalence of SARS-CoV-2 in children has increased, with notable regional and demographic disparities. COVID-19-related hospitalisation rates in children rose during Omicron waves, especially among infants, unvaccinated individuals, and CYP at higher risk, i.e. with comorbidities such as obesity, diabetes, and neurological or cardiac conditions. Despite this, severe disease and mortality in children remained very low. The observed increases in type 1 diabetes incidence and multisystem inflammatory syndrome in children (MIS-C) have also highlighted the broader systemic effects of SARS-CoV-2 in paediatric populations. Evidence has underscored the protective effect of vaccination in preventing severe disease and MIS-C and vaccine safety, emphasising the need for targeted immunisation strategies, particularly among children who may be at higher risk. Studies have also estimated that a significant proportion of children experienced persistent post-COVID-19 infection symptoms such as fatigue, mood disturbances, sleep disorders, and respiratory difficulties, but the reported prevalence varied widely, from as low as 1.6% to as high as 70%, due to differences in study methodologies, case definitions, and populations studied. Standardised definitions and measurement tools, such as those developed through international consensus processes, are required to improve diagnosis, treatment, and research into this persisting condition. Ethnic disparities in vaccine uptake persist, implying that vaccine hesitancy and accessibility, alongside approaches to countering disinformation, are important areas for future research.
Collapse
Affiliation(s)
- Igor Rudan
- Usher Institute, The University of Edinburgh, Edinburgh, UK
- Nuffield Department of Primary Care Health Sciences, Oxford University, UK
| | - Steven Kerr
- Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - Colin R Simpson
- Usher Institute, The University of Edinburgh, Edinburgh, UK
- School of Health, Wellington Faculty of Health, Victoria University of Wellington, Wellington, New Zealand
| | - Amanj Kurdi
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Strathclyde University, Glasgow, UK
- College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq
- College of Pharmacy, Al-Kitab University, Kirkuk, Iraq
- Department of Public Health Pharmacy and Management, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| | | | - Chris Robertson
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, UK
- Public Health Scotland, Glasgow, UK
| | - Aziz Sheikh
- Usher Institute, The University of Edinburgh, Edinburgh, UK
- Nuffield Department of Primary Care Health Sciences, Oxford University, UK
| | - EAVE II Collaboration
- Usher Institute, The University of Edinburgh, Edinburgh, UK
- Nuffield Department of Primary Care Health Sciences, Oxford University, UK
- School of Health, Wellington Faculty of Health, Victoria University of Wellington, Wellington, New Zealand
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Strathclyde University, Glasgow, UK
- College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq
- College of Pharmacy, Al-Kitab University, Kirkuk, Iraq
- Department of Public Health Pharmacy and Management, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, South Africa
- Teesside University, Middlesborough, UK
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, UK
- Public Health Scotland, Glasgow, UK
| |
Collapse
|
7
|
Santa Ardisson J, Vedovatti Monfardini Sagrillo M, Ramos Athaydes B, Corredor Vargas AM, Torezani R, Ribeiro-Rodrigues R, Cruz Spano L, Gaburro Paneto G, Delatorre E, Ventorin von Zeidler S, Freire Bastos Filho T. Comparative spatial-temporal analysis of SARS-CoV-2 lineages B.1.1.33 and BQ.1.1 Omicron variant across pandemic phases. Sci Rep 2025; 15:10319. [PMID: 40133656 PMCID: PMC11937565 DOI: 10.1038/s41598-025-95140-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 03/19/2025] [Indexed: 03/27/2025] Open
Abstract
The evolution of COVID-19 pandemic has been characterized by the rapid emergence of new SARS-CoV-2 variants, each of which poses unique challenges to public health. This study analyzes the dispersion profiles during the Pre-Omicron and Post-Omicron phases in different epidemiological contexts. The Brazilian state of Espirito Santo, despite its low population density, plays a critical role as a commercial hub due to its intense port activity, which may have contributed to COVID-19 cases and mortality rates being higher than the national average. The state recorded 34,000 confirmed cases and 377 deaths per 100,000 inhabitants. Genomic surveillance revealed that the Pre-Omicron phase was dominated by the B.1.1.33 lineage, characterized by localized intraregional circulation. In contrast, the Post-Omicron phase, dominated by the BQ.1.1 lineage, exhibited greater diversity in circulating lineages, increased international interactions, and rapid viral dissemination, highlighting distinct transmission dynamics between such periods. This study highlights the need for adaptive public health strategies that account for both viral behavior and regional socioeconomic factors, while highlighting the strategic importance of Espirito Santo in monitoring SARS-CoV-2 evolution.
Collapse
Affiliation(s)
- Juliana Santa Ardisson
- Postgraduate Program in Biotechnology, Federal University of Espirito Santo, Vitoria, 29047-105, Brazil.
| | | | - Brena Ramos Athaydes
- Postgraduate Program in Biotechnology, Federal University of Espirito Santo, Vitoria, 29047-105, Brazil
- Department of Pathology, Federal University of Espirito Santo, Vitoria, 29047-105, Brazil
| | | | - Renata Torezani
- Postgraduate Program in Biotechnology, Federal University of Espirito Santo, Vitoria, 29047-105, Brazil
| | - Rodrigo Ribeiro-Rodrigues
- Postgraduate Program in Infectious Diseases, Federal University of Espirito Santo, Vitoria, 29047-105, Brazil
- Department of Pathology, Federal University of Espirito Santo, Vitoria, 29047-105, Brazil
- Central Public Health Laboratory of the State of Espirito Santo (LACEN-ES), Vitoria, 29050-260, Brazil
| | - Liliana Cruz Spano
- Postgraduate Program in Infectious Diseases, Federal University of Espirito Santo, Vitoria, 29047-105, Brazil
| | - Greiciane Gaburro Paneto
- Postgraduate Program in Biotechnology, Federal University of Espirito Santo, Vitoria, 29047-105, Brazil
| | - Edson Delatorre
- Postgraduate Program in Infectious Diseases, Federal University of Espirito Santo, Vitoria, 29047-105, Brazil
- Department of Pathology, Federal University of Espirito Santo, Vitoria, 29047-105, Brazil
| | - Sandra Ventorin von Zeidler
- Postgraduate Program in Biotechnology, Federal University of Espirito Santo, Vitoria, 29047-105, Brazil
- Department of Pathology, Federal University of Espirito Santo, Vitoria, 29047-105, Brazil
| | | |
Collapse
|
8
|
Malacatus-Arboleda A, Barbotó-Ramírez E, Sánchez GE, Moscoso B, Rhodes LA, Coloma J, Guevara Á, Espinoza-Fuentes F, Fernández-Cadena JC, Morey-León G, Andrade-Molina D. The effect of COVID vaccination timing on the seroprevalence of IgG antibodies: evidence from the Guayas region of Ecuador. Front Public Health 2025; 13:1537049. [PMID: 40201360 PMCID: PMC11975873 DOI: 10.3389/fpubh.2025.1537049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
Background and aims Timely distribution of COVID-19 vaccines was particularly important for developing countries that do not have strong health systems and related infrastructure. We analyze data from the Guayas province of Ecuador, an area particularly affected by the pandemic, to determine the seroprevalence of SARS-CoV-2 and the effect of the timing of the second dose of COVID-19 vaccines on the seroprevalence SARS-CoV-2 IgG antibodies. Methods This cross-sectional study involved 1,761 individuals aged 18 and older who voluntarily enrolled prior to and during the initial phase of vaccine rollout in Ecuador (October 2020 to July 2022). IgG anti-SARS-CoV-2 RBD antibodies were assessed by an in-house ELISA to evaluate the immune response to Pfizer (BioNTech, Spike mRNA) and AstraZeneca (Oxford, AstraZeneca Spike) vaccine in the Guayas province. Ordinary least squares (OLS) regressions were employed to determine the effect of delayed second doses later than prescribed by the manufacturer for both vaccines. Results Before the vaccination campaign, we estimated an RBD IgG seroprevalence of 27.7% (95% CI: 23.6-27, n = 469). The estimate increased to 89.4% (95% CI: 87.7-91.18, n = 1,235) after the first vaccine dose and to 92.6% (95% CI: 90.7-94.5, n = 748) after the second dose. Individuals who received the second dose of the Pfizer vaccine later than the recommended dose showed significantly lower levels of IgG antibodies 2-3 weeks after receiving the second dose than those who received the dose within the recommended timeframe. Furthermore, we did not find any effect on RBD IgG antibody levels in those who received a second dose of the AstraZeneca vaccine during the first and second parts of the recommended vaccination window. Conclusion The results suggest that a significant portion of the study population was already infected with SARS-CoV-2 prior to the vaccination. As expected, seropositivity increased alongside vaccination efforts. We determined that Pfizer vaccine recipients should be adhered to vaccine timing guidelines. Furthermore, resource-limited countries should consider administering vaccines with flexibility in dosing intervals, such as AstraZeneca, as it allows for a wider time frame without significantly reducing the boosting of IgG antibodies.
Collapse
Affiliation(s)
- Aurora Malacatus-Arboleda
- Laboratorio de Ciencias Ómicas, Facultad de Ciencias de la Salud, Universidad Espíritu Santo, Samborondón, Ecuador
| | - Erick Barbotó-Ramírez
- Laboratorio de Ciencias Ómicas, Facultad de Ciencias de la Salud, Universidad Espíritu Santo, Samborondón, Ecuador
| | - Gonzalo E. Sánchez
- Facultad de Ciencias Sociales y Humanísticas, Centro de Investigaciones Económicas, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Bernard Moscoso
- Facultad de Ciencias Sociales y Humanísticas, Centro de Investigaciones Económicas, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Lauren A. Rhodes
- Facultad de Ciencias Sociales y Humanísticas, Centro de Investigaciones Rurales, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Josefina Coloma
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, United States
| | - Ángel Guevara
- Instituto de Biomedicina, Carrera de Medicina, Universidad Central, Quito, Ecuador
| | - Fernando Espinoza-Fuentes
- Laboratorio de Ciencias Ómicas, Facultad de Ciencias de la Salud, Universidad Espíritu Santo, Samborondón, Ecuador
| | - Juan Carlos Fernández-Cadena
- Laboratorio de Ciencias Ómicas, Facultad de Ciencias de la Salud, Universidad Espíritu Santo, Samborondón, Ecuador
- Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, United States
| | - Gabriel Morey-León
- Laboratorio de Ciencias Ómicas, Facultad de Ciencias de la Salud, Universidad Espíritu Santo, Samborondón, Ecuador
| | - Derly Andrade-Molina
- Laboratorio de Ciencias Ómicas, Facultad de Ciencias de la Salud, Universidad Espíritu Santo, Samborondón, Ecuador
| |
Collapse
|
9
|
Adamczuk J, Kamiński KA, Sołomacha S, Kazberuk M, Chlabicz M, Czupryna P, Dunaj-Małyszko J, Citko A, Sowa P, Dubatówka M, Łapińska M, Kiszkiel Ł, Szczerbiński Ł, Laskowski PP, Alimowski M, Moniuszko-Malinowska A. Determinants and dynamics of the seroprevalence of anti-SARS-CoV-2 antibodies in Poland. Adv Med Sci 2025; 70:219-228. [PMID: 40097104 DOI: 10.1016/j.advms.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/08/2024] [Accepted: 03/11/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE In the present study, we aimed to assess the anti-SARS-CoV-2 antibodies in population and in convalescents 6 months after infection with COVID-19. PATIENTS AND METHODS The study population consisted of 2 groups, i.e. group I: 232 post-COVID-19 patients, and group II: 544 patients from a population cohort. Detection of anti-N antibodies was performed with Elecsys Anti-SARS-CoV-2 and anti-S antibodies with LIAISON SARS-CoV-2 S1/S2 IgG tests. RESULTS After the Omicron wave, gradual increase in the prevalence of the analyzed antibodies was observed in the general population - up to 92.5 % of anti-S and 69.7 % of anti-N antibodies. In the COVID-19 convalescents, 6 months after infection, 4.3 % and 3.7 % did not generate significant titers of anti-S and anti-N antibodies, respectively. Among patients, who were vaccinated, 1 % did not generate anti-S antibodies. The median age of non-responders was higher than that of responders. Sex did not influence the results. Comparison of anti-N and anti-S antibodies 6 months after infection in the whole group showed higher anti-N titer in patients who suffered from the disease than in the general population. In the general population, 17.4 % of participants had anti-N antibodies, and 9 % had anti-S antibodies, despite no prior vaccination or history of COVID-19. CONCLUSIONS High prevalence of anti-N antibodies in patients reporting no history of COVID-19 during last wave, even after official end of the pandemic suggests high prevalence of SARS-CoV-2 in the environment and frequent asymptomatic or unspecific COVID-19, what is of high importance from the public health perspective.
Collapse
Affiliation(s)
- Justyna Adamczuk
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| | - Karol Adam Kamiński
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Sebastian Sołomacha
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Małgorzata Kazberuk
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Magdalena Chlabicz
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Piotr Czupryna
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| | - Justyna Dunaj-Małyszko
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| | - Anna Citko
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Paweł Sowa
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Marlena Dubatówka
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Magda Łapińska
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Łukasz Kiszkiel
- Society and Cognition Unit, University of Bialystok, Bialystok, Poland
| | - Łukasz Szczerbiński
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland; Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | | | - Maciej Alimowski
- Doctoral School of Social Sciences, University of Bialystok, Bialystok, Poland
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
10
|
Tembo T, Heilmann E, Kabamba BM, Fwoloshi S, Kalenga K, Chilambe F, Siwinga M, Rutagwera MR, Musunse M, Kangale C, Yingst S, Yadav R, Savory T, Gutman JR, Sikazwe I, Mulenga LB, Moore CB, Hines JZ. Implementing SARS-CoV-2 routine surveillance in antenatal care in Zambia, 2021-2022: best practices and lessons learned. BMC Public Health 2025; 25:813. [PMID: 40021963 PMCID: PMC11869463 DOI: 10.1186/s12889-025-21918-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 02/12/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND In Zambia, the true extent of SARS-CoV-2 infections is unknown because initial surveillance focused on patients with symptoms or severe disease. Antenatal sentinel surveillance had not been used to assess infection trends. The ANC COVID-19 surveillance study sought to determine SARS-CoV-2 seroprevalence and COVID-19 vaccine uptake among pregnant women. We provide insight into the study implementation, challenges encountered, best practices, and lessons learned. METHODS A repeated cross-sectional seroprevalence survey was implemented at 39 health facilities in four districts from September 2021 to September 2022. Pregnant women aged 15-49 years were enrolled at their first antenatal care visits. An electronic questionnaire gathered demographics and other COVID-19 related information from consenting participants. A dried blood sample was collected to detect IgG antibodies using a multiplex bead assay. Seropositive results were categorized as infection, infection and vaccination or infection based on anti-RBD and anti-nucleocapsid test results. Problems and their root causes were identified as they occurred. Practical problem-solving strategies were devised, implemented, and monitored to ensure that goals were accomplished. RESULTS In the primary analysis, 7% of the 9,221 samples collected from participants were not tested because they were missing. COVID-19 vaccine uptake of 9,111 pregnant women was assessed. Approximately 64% of participants were cumulatively seropositive for SARS-CoV-2 antibodies. Seroprevalence increased from 27.8% in September 2021 to 56.6% in July 2022. We observed an increase in vaccine coverage (0.5-27%) over time. Women aged 40-49 years old, without education and with prior COVID-19 infection were associated with higher vaccine uptake. The Delta variant of COVID-19 and the reallocation of health facilities between two partners delayed surveillance activities and increased the cost of implementation (e.g., the purchase of additional calibration and validation kits and DBS cards). Protocol deviations were attributed to the lack of experience in conducting research but, the district RAs repeatedly trained health facility staff to enhance their research knowledge. CONCLUSIONS Incorporating SARS-CoV-2 surveillance into routine antenatal care is feasible and potentially sustainable when existing health system infrastructure, human resources, and surveillance systems are leveraged. Yet, careful planning is needed to anticipate implementation challenges and ensure high-quality data collection.
Collapse
Affiliation(s)
- Tannia Tembo
- Centre for Infectious Disease Research in Zambia (CIDRZ), P.O Box 34681, Lusaka, Zambia.
| | - Elizabeth Heilmann
- Public Health Institute, Oakland, CA, USA
- U.S. Centers for Disease Control and Prevention, Lusaka, Zambia
| | | | | | - Kalubi Kalenga
- Centre for Infectious Disease Research in Zambia (CIDRZ), P.O Box 34681, Lusaka, Zambia
| | | | | | | | | | | | - Samuel Yingst
- U.S. Centers for Disease Control and Prevention, Lusaka, Zambia
| | - Ruchi Yadav
- U.S. Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Theodora Savory
- Centre for Infectious Disease Research in Zambia (CIDRZ), P.O Box 34681, Lusaka, Zambia
| | - Julie R Gutman
- U.S. Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Izukanji Sikazwe
- Centre for Infectious Disease Research in Zambia (CIDRZ), P.O Box 34681, Lusaka, Zambia
| | | | - Carolyn Bolton Moore
- Centre for Infectious Disease Research in Zambia (CIDRZ), P.O Box 34681, Lusaka, Zambia
- University of Alabama at Birmingham, Tuscaloosa, USA
| | - Jonas Z Hines
- U.S. Centers for Disease Control and Prevention, Lusaka, Zambia
| |
Collapse
|
11
|
Bhiman JN, Madzorera VS, Mkhize Q, Scheepers C, Hermanus T, Ayres F, Makhado Z, Moyo-Gwete T, Crowther C, Singh B, Fortuin M, Marinda E, Jooste S, Zuma K, Zungu N, Morris L, Puren A, Simbayi L, Moyo S, Moore PL. Population shift in antibody immunity following the emergence of a SARS-CoV-2 variant of concern. Sci Rep 2025; 15:5549. [PMID: 39953108 PMCID: PMC11828959 DOI: 10.1038/s41598-025-89940-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) exhibit escape from pre-existing immunity and elicit variant-specific immune responses. In South Africa, the second wave of SARS-CoV-2 infections was driven by the Beta VOC, which coincided with the country-wide National COVID-19 Antibody Survey (NCAS). The NCAS was conducted between November 2020 and February 2021 to understand the burden of SARS-CoV-2 infection through seroprevalence. We evaluated 649 NCAS sera for spike binding and pseudovirus neutralizing antibodies. We classified individuals as ancestral or D614G neutralizers (114/649), Beta neutralizers (96/649), double neutralizers (375/649) or non-neutralizers (62/649). We observed a consistent decrease in preferential neutralization against the D614G variant from 68 to 18% of individuals over the four sampling months. Concurrently, samples with equivalent neutralization of both variants, or with enhanced neutralization of the Beta variant, increased from 32 to 82% of samples. Neutralization data showed that geometric mean titers (GMTs) against D614G dropped 2.4-fold, while GMTs against Beta increased 2-fold during this same period. A shift in population humoral immunity in favor of Beta-directed or cross-neutralizing antibody responses, paralleled the increase in genomic frequency of the Beta variant in South Africa. Understanding similar population immunity shifts could elucidate immunity gaps that drive SARS-CoV-2 evolution.
Collapse
Affiliation(s)
- Jinal N Bhiman
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa.
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa.
| | - Vimbai Sharon Madzorera
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Qiniso Mkhize
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Cathrine Scheepers
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Tandile Hermanus
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Frances Ayres
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Zanele Makhado
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Thandeka Moyo-Gwete
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Carol Crowther
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Beverley Singh
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Mirriam Fortuin
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Edmore Marinda
- Human Sciences Research Council, Pretoria, South Africa
- School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Sean Jooste
- Human Sciences Research Council, Pretoria, South Africa
| | - Khangelani Zuma
- Human Sciences Research Council, Pretoria, South Africa
- School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Nompumelelo Zungu
- Human Sciences Research Council, Pretoria, South Africa
- School of Nursing and Public Health, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Lynn Morris
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Adrian Puren
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Leickness Simbayi
- Human Sciences Research Council, Pretoria, South Africa
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Sizulu Moyo
- Human Sciences Research Council, Pretoria, South Africa
| | - Penny L Moore
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa.
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa.
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa.
| |
Collapse
|
12
|
Bergna A, Lai A, Sagradi F, Menzo S, Mancini N, Bruzzone B, Rusconi S, Marchegiani G, Clementi N, Francisci D, Vicenti I, Ronchiadin S, Mbissam HD, della Ventura C, Lanfranchi L, Testa S, Caucci S, Acciarri C, Carioti L, Occhionero A, Novazzi F, Genoni AP, Ferrante FD, De Pace V, Ferraris M, Ogliastro M, Gabrieli A, De Paschale M, Canavesi G, Bellocchi MC, Iannetta M, Sarmati L, Ceccherini‐Silberstein F, Riva A, Antinori S, Zehender G. Genomic Epidemiology of the Main SARS-CoV-2 Variants Circulating in Italy During the Omicron Era. J Med Virol 2025; 97:e70215. [PMID: 39936851 PMCID: PMC11816846 DOI: 10.1002/jmv.70215] [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: 07/25/2024] [Revised: 01/09/2025] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
Since early 2022 the Omicron variant has rapidly spread worldwide, becoming the dominant variant to date. The study aimed to investigate the clinical and epidemiological characteristics of COVID-19 patients and reconstruct the genomic epidemiology of main SARS-CoV-2 Omicron sublineages in Italy in 2022. A total of 8970 SARS-CoV-2 samples were studied, and phylogenetic analyses were focused on BA.1, BA.2, and BA.5 subvariants. More than half of subjects received three doses of vaccine and experienced a reinfection. A significant larger proportion of unvaccinated subjects presented reinfection compared with vaccinated. Clusters presented a tMRCA between September-November 2021 (BA.1), November 2021-January 2022 (BA.2), and October 2021-May 2022 (BA.5). Re values showed the highest level between September-October, January-February 2022, and May 2022 for BA.1, BA.2 and BA.5, respectively. Limited number of studied variant sequences are included in clusters. The spread rate of the studied variant exceeded its evolutionary rate. No single sublineage had sufficient time to differentiate into large clusters, but only into small and fragmented groups sharing the same recent ancestor. These analyses dissect the epidemiological dynamics of Omicron sublineages in Italy over a period of great epidemiological changes in the COVID-19 epidemic.
Collapse
Affiliation(s)
- Annalisa Bergna
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | - Alessia Lai
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | - Fabio Sagradi
- Unit of Infectious DiseasesAzienda Socio‐Sanitaria Territoriale CremonaCremonaItaly
| | - Stefano Menzo
- Department of Biomedical Sciences and Public HealthVirology Unit, Polytechnic University of MarcheAnconaItaly
| | - Nicasio Mancini
- Department of Medicine and Technological InnovationUniversity of InsubriaVareseItaly
- Laboratory of Medical Microbiology and VirologyOspedale di Circolo e Fondazione MacchiVareseItaly
| | | | - Stefano Rusconi
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
- Ospedale Civile di Legnano ASST Ovest MilaneseUniversity of MilanLegnanoItaly
| | - Greta Marchegiani
- Department of Experimental MedicineUniversity of Rome “Tor Vergata”RomeItaly
| | - Nicola Clementi
- Laboratory of Microbiology and VirologyUniversità “Vita‐Salute” San RaffaeleMilanItaly
- IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Daniela Francisci
- Department of Medicine and Surgery, Clinic of Infectious Diseases“Santa Maria della Misericordia” Hospital, University of PerugiaPerugiaItaly
| | - Ilaria Vicenti
- Department of Medical BiotechnologiesUniversity of SienaSienaItaly
| | - Silvia Ronchiadin
- Intesa Sanpaolo Innovation Center‐ Artificial Intelligence LabTurinItaly
| | | | | | - Leonardo Lanfranchi
- Unit of Infectious DiseasesAzienda Socio‐Sanitaria Territoriale CremonaCremonaItaly
| | - Sophie Testa
- Unit of Infectious DiseasesAzienda Socio‐Sanitaria Territoriale CremonaCremonaItaly
| | - Sara Caucci
- Department of Biomedical Sciences and Public HealthVirology Unit, Polytechnic University of MarcheAnconaItaly
| | - Carla Acciarri
- Department of Biomedical Sciences and Public HealthVirology Unit, Polytechnic University of MarcheAnconaItaly
| | - Luca Carioti
- Department of Experimental MedicineUniversity of Rome “Tor Vergata”RomeItaly
| | | | - Federica Novazzi
- Department of Medicine and Technological InnovationUniversity of InsubriaVareseItaly
- Laboratory of Medical Microbiology and VirologyOspedale di Circolo e Fondazione MacchiVareseItaly
| | - Angelo Paolo Genoni
- Department of Medicine and Technological InnovationUniversity of InsubriaVareseItaly
- Laboratory of Medical Microbiology and VirologyOspedale di Circolo e Fondazione MacchiVareseItaly
| | - Francesca Drago Ferrante
- Department of Medicine and Technological InnovationUniversity of InsubriaVareseItaly
- Laboratory of Medical Microbiology and VirologyOspedale di Circolo e Fondazione MacchiVareseItaly
| | | | | | | | - Arianna Gabrieli
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
- Unit of MicrobiologyLegnano Hospital, ASST Ovest MilaneseLegnanoItaly
| | | | - Giada Canavesi
- Ospedale Civile di Legnano ASST Ovest MilaneseUniversity of MilanLegnanoItaly
| | | | - Marco Iannetta
- Department of System Medicine, Clinical Infectious DiseasesTor Vergata UniversityRomeItaly
| | - Loredana Sarmati
- Department of System Medicine, Clinical Infectious DiseasesTor Vergata UniversityRomeItaly
| | | | - Agostino Riva
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | - Spinello Antinori
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | | |
Collapse
|
13
|
Figueroa AL, Torres D, Reyes-Acuna C, Matherne P, Yeakey A, Deng W, Xu W, Sigal Y, Chambers G, Olsen M, Girard B, Miller JM, Das R, Priddy F. Safety and immunogenicity of a single-dose omicron-containing COVID-19 vaccination in adolescents: an open-label, single-arm, phase 2/3 trial. THE LANCET. INFECTIOUS DISEASES 2025; 25:208-217. [PMID: 39332418 DOI: 10.1016/s1473-3099(24)00501-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND Most individuals show immunity to SARS-CoV-2 from vaccination or infection, or both. We aimed to determine the safety and immunogenicity of an omicron-containing COVID-19 vaccine (mRNA-1273.222) in vaccine-naive adolescents who were SARS-CoV-2 positive. METHODS Part 3 of the phase 2/3 TeenCOVE trial was a phase 3, open-label, single-arm part done in the USA and the Dominican Republic that enrolled healthy, vaccine-naive adolescents (aged 12-17 years) to receive two 50 μg doses of mRNA-1273.222 (ancestral strain Wuhan-Hu-1 and omicron subvariants BA.4 and BA.5), 6 months apart. Primary reactogenicity and safety outcomes included assessment of solicited local or systemic adverse reactions 7 days after vaccination, and unsolicited and prespecified adverse events throughout study participation. Inferred effectiveness (primary immunogenicity outcome) was established by comparing neutralising antibody responses 28 days after dose 1 of mRNA-1273.222 in SARS-CoV-2-positive adolescents with responses 28 days after dose 2 of mRNA-1273 100 μg primary series in SARS-CoV-2-negative young adults (aged 18-25 years) from the COVE trial. This study is registered with ClinicalTrials.gov (NCT04649151). FINDINGS Between Dec 21, 2022, and June 5, 2023, 379 adolescents (378 of whom were SARS-CoV-2 positive) received at least one mRNA-1273.222 dose and were included in the safety analysis set. The reactogenicity profile was favourable compared with the mRNA-1273 primary series, with no new safety concerns identified. Unsolicited adverse events were reported in 49 (13%) of 379 participants; no deaths or adverse events leading to study discontinuation were reported. The immunogenicity set included 245 adolescents from the per-protocol immunogenicity subset who were SARS-CoV-2 positive at baseline and 296 young adults who were SARS-CoV-2 negative. Compared with the mRNA-1273 primary series in SARS-CoV-2-negative young adults, a single dose of mRNA-1273.222 induced superior (geometric mean ratio [GMR] 95% CI lower bound >1) neutralising antibody responses against omicron BA.4 and BA.5 (GMR 48·95 [95% CI 44·21-54·21]) and non-inferior (GMR 95% CI lower bound >0·667) neutralising antibody responses against ancestral SARS-CoV-2 (GMR 4·25 [95% CI 3·69-4·88]) in SARS-CoV-2-positive adolescents. INTERPRETATION In vaccine-naive, SARS-CoV-2-positive adolescents, single-dose mRNA-1273.222 was effective against COVID-19 based on successful immunobridging to the two-dose mRNA-1273 primary series in young adults. The findings support a simplified single-dose vaccination schedule with variant-containing mRNA vaccines, regardless of previous vaccination status. FUNDING Moderna.
Collapse
Affiliation(s)
| | - Dania Torres
- Hospital General Regional Dr Marcelino Velez Santana, Santo Domingo, Dominican Republic
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Sturrock S, Cavell B, Alexander F, Apostolakis K, Barro C, Daniel O, Dixon L, Halkerston R, Hall T, Hesp JR, Hill AM, Leung S, Lim S, McStraw N, Otter A, Ramkhelawon L, Watts R, Etti M, Heath P, Lee-Wo C, Greening V, Khalil A, Turner K, Taylor S, Doare KL, Ladhani S. Maternal and Placental Antibody Responses in SARS-CoV-2 Vaccination and Natural Infection During Pregnancy. Pediatr Infect Dis J 2025; 44:S32-S37. [PMID: 39951071 PMCID: PMC7617455 DOI: 10.1097/inf.0000000000004704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
BACKGROUND As COVID-19 becomes endemic, understanding antibody response and transfer during pregnancy is crucial to inform policy and vaccination schedules. While good immunogenicity has been shown from SARS-CoV-2 vaccines, few data are available demonstrating functional responses in pregnant populations and infants. METHODS A prospective, multi-site observational study was completed across 14 centers in England from April 23, 2020, to December 21, 2022. Demographic, COVID infection and vaccination data were collected. Maternal and cord blood samples were taken at delivery, with maternal and neonatal blood samples taken at 6 weeks for participants who had been infected or vaccinated. Antibody concentrations were measured using antibody-dependent complement deposition, antibody-dependent neutrophil phagocytosis, ACE2 inhibition and Roche and EuroImmun antibody binding assays at the UK Health Security Agency. RESULTS Maternal vaccination and infection both produced an antibody response in 100% of mothers and 93.8% and 92.9% of neonates, respectively, which persisted at 6 weeks in 95%. The strongest response was seen in mothers who were both vaccinated and infected. Anti-spike antibody response decreased almost 25-fold from first to third trimester vaccination (P=0.013). Placental transfer of antibodies post-infection showed varied results depending on the assay used, with higher transfer ratios observed in assays measuring Fc-mediated antibody effector functions and IgG-specific responses. CONCLUSIONS Maternal vaccination is associated with good immunogenicity and successful antibody transfer to the neonate, particularly with vaccination in early pregnancy. Further study is needed to determine the mechanism by which the timing of vaccination affects antibody transfer. When measuring placental transfer of antibodies, consideration of the assay to use is essential.
Collapse
Affiliation(s)
- Sarah Sturrock
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | | | | | | | - Camille Barro
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | - Olwenn Daniel
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | | | | | - Tom Hall
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | | | | | | | - Suzy Lim
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | | | | | - Laxmee Ramkhelawon
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | - Robert Watts
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | - Melanie Etti
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | - Paul Heath
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | - Chelone Lee-Wo
- St George’s Hospital NHS Trust, UK
- St George’s, University of London, UK
| | - Vanessa Greening
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| | | | - Kim Turner
- Keck School of Medicine, University of Southern California
| | | | - Kirsty Le Doare
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
- Makerere University-Johns Hopkins University Research Collaboration, Uganda
| | - Shamez Ladhani
- Centre for Neonatal and Paediatric Infection, St George’s, University of London, UK
| |
Collapse
|
15
|
Blom K, Galanis I, Bacchus P, Sondén K, Bujila I, Efimova T, Garli F, Mansjö M, Movert E, Pettke A, Rapp M, Sperk M, Söderholm S, Valentin Asin K, Zanetti S, Gisslén M, Bråve A, Groenheit R, Klingström J. High prevalence of undocumented SARS-CoV-2 infections revealed by analysis of nucleocapsid-specific IgG responses in diagnosed and undiagnosed individuals. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0003300. [PMID: 39841747 PMCID: PMC11753678 DOI: 10.1371/journal.pgph.0003300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 12/05/2024] [Indexed: 01/24/2025]
Abstract
Acute SARS-CoV-2 infections are not always diagnosed; hence an unknown proportion of all infections are not documented. SARS-CoV-2 can induce spike and nucleocapsid protein specific IgG antibodies, which can be detected in seroprevalence studies to identify a previous infection. However, with the introduction of vaccines containing the spike protein it is no longer possible to use spike-IgG as a marker of infection. In many countries marketed vaccines do not include the nucleocapsid protein, allowing the use of nucleocapsid-specific IgG (N-IgG) as a specific marker for previous infection. Importantly however, not all SARS-CoV-2-infected individuals develop detectable N-IgG responses and there are reports of waning of N-IgG titers in previously infected individuals, complicating the use of N-IgG in seroprevalence studies. Here, our aim was to investigate N-IgG as a marker for previous infection. To this end we analyzed a well characterized cohort (n = 2,583; sampled in March, 2022), including 612 participants with a previously diagnosed and documented SARS-CoV-2-infection. We show that 75% (460/612) of the confirmed SARS-CoV-2-infected participants were N-IgG positive, and that the frequency of seropositivity was stable for at least 105 weeks after the latest documented SARS-CoV-2-infection. Among participants with no documented SARS-CoV-2-infection, 32.6% (642/1971) were N-IgG-positive, suggesting a previous infection. Assuming similar frequency of N-IgG-seronegative cases in previously diagnosed and undiagnosed individuals we further estimate that 214 of the 1329 undiagnosed and N-IgG-negative cases had been previously infected, indicating a total infection rate of 56.8% (1,468/2,583), clearly higher than the documented 23.7% rate of infection, in this cohort. In conclusion, our results suggest that while N-IgG is a good marker of previous SARS-CoV-2-infection the large proportion of previously infected N-IgG-negative individuals introduces a risk for underestimations of total level of previously infected individuals in a population. Accounting for this dark number of undiagnosable cases can provide better estimates of total level of infected individuals in a population.
Collapse
Affiliation(s)
- Kim Blom
- Public Health Agency of Sweden, Solna, Sweden
| | | | - Philip Bacchus
- Lund University, Lund, Sweden
- Swedish Armed Forces, Umeå, Sweden
| | | | | | | | | | | | - Elin Movert
- Public Health Agency of Sweden, Solna, Sweden
| | | | - Marie Rapp
- Public Health Agency of Sweden, Solna, Sweden
| | - Maike Sperk
- Public Health Agency of Sweden, Solna, Sweden
| | | | | | | | - Magnus Gisslén
- Public Health Agency of Sweden, Solna, Sweden
- University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | | | - Jonas Klingström
- Public Health Agency of Sweden, Solna, Sweden
- Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
16
|
Menezes A, Razafimahatratra SL, Wariri O, Graham AL, Metcalf CJE. Strengthening serological studies: the need for greater geographical diversity, biobanking, and data-accessibility. Trends Microbiol 2025:S0966-842X(24)00322-6. [PMID: 39818508 DOI: 10.1016/j.tim.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
Serological studies uniquely strengthen infectious disease surveillance, expanding prevalence estimates to encompass asymptomatic infections, and revealing the otherwise inapparent landscape of immunity, including who is and is not susceptible to infection. They are thus a powerful complement to often incomplete epidemiological and public health measures (administrative measures of vaccination coverage, incidence estimates, etc.). The recent surge in the deployment of serological surveys globally (in part due to the SARS-CoV-2 pandemic), alongside the development of new assays and new inference methods, means that the time is ripe to interrogate areas to strengthen future serosurveillance efforts. We identify three themes warranting attention: first, expanding the geographical diversity of these studies; second, investing globally in infrastructure for storage of blood samples (biobanking), opening the way to future analyses; and third, establishing protocols to increase data accessibility and to facilitate data usage for current and future studies. We conclude that strengthening serological studies is necessary and achievable through thoughtful sampling design, wide-scale sample storage, and thorough reporting practices.
Collapse
Affiliation(s)
- Arthur Menezes
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA.
| | | | - Oghenebrume Wariri
- Vaccines and Immunity Theme, MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, London, Banjul, The Gambia; Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Andrea L Graham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA; Santa Fe Institute, Santa Fe, NM, USA
| | - C Jessica E Metcalf
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA; Princeton School of Public and International Affairs, Princeton University, Princeton, NJ, USA
| |
Collapse
|
17
|
Fachiroh J, Lestari SK, Paramita DK, Bintoro BS, Dewi FST, Lazuardi L, Rusadi CP, Sagenah EN, Arguni E. Seroconversion and dynamics of IgG anti-SARS-CoV-2 antibodies during the pandemic: A two-month observation cohort study on the population of Sleman in Indonesia. PLoS One 2025; 20:e0316360. [PMID: 39746050 PMCID: PMC11695021 DOI: 10.1371/journal.pone.0316360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND This study describes the seroconversion and serodynamics of IgG antibodies against the RBD of SARS-CoV-2 in the general population of Sleman District, Yogyakarta Special Province. We aim to identify possible factors that correlate with the seroconversion and serodynamics of IgG antibodies against the RBD of SARS-CoV-2. METHODS We performed a longitudinal study of the population at Health and Demographic Surveillance System (HDSS) Sleman, Yogyakarta, Indonesia. Study subjects were recruited between April and December 2021 using convenience sampling and were followed up 2 times, i.e. 4-5 and 8-9 weeks. The inclusion criteria for subjects were age ≥ 18 years, absence of flu-like symptoms, and negative COVID-19 by using GeNose C19® screening. A community-based survey on demographics, comorbidities and smoking habits were documented at baseline, while a history of vaccination, COVID-19-related symptoms, mobility, and preventive measures, weight and height as well as a venous blood draw, were collected at each visit. The anti-RBD-SARS-CoV-2 IgG antibody concentration from blood plasma was measured using chemiluminescent microplate immunoassay (CMIA). Descriptive analysis was performed based on IgG seropositivity by using chi-squared test or Fisher's exact test, as appropriate. Logistic regression was subsequently performed to identify factors that were correlated with IgG seropositivity. Further, a grouping of subjects based on IgG seropositivity was done to analyze factors that might correlate with seroconversion and serodynamics of anti-RBD-SARS-CoV-2 IgG antibody. A P value ≤ 0.05 was considered to indicate a significant difference. RESULTS Three hundred eighty-five (385) participants were analyzed. At baseline, 307 out of 385 (79.7%) subjects were seropositive for the IgG antibody against the RBD of SARS-CoV-2. Descriptive analysis showed that sex, marital status, smoking habits, obesity, vaccination status, and preventive measures were different between the IgG anti-RBD-SARS-CoV-2 seropositive and negative individuals (p≤ 0.05). Further analysis showed that, vaccination was the factor most strongly correlated with seropositivity [OR = 20.58; 95% CI 10.82, 39.15]. Based on the correlation, we separated subjects into 4 groups. Group 1 (seronegative-unvaccinated individuals; 50 subjects); Group 2 (seronegative-vaccinated individuals; 27 subjects); Group 3 (seropositive-unvaccinated individuals; 25 subjects); and Group 4 (seropositive-vaccinated individuals; 282 subjects). During monitoring, 27/49 (55.10%), 5/25 (20%), 9/22 (40.91%), and 27/257 (10.51%) of subjects in Group 1, 2, 3, and 4 respectively, received 1 or 2 doses of COVID19 vaccine. When comparing seroconversion at baseline and monitoring 2, positive IgG seroconversion was observed in Group 1 (from 0/51 (0%) to 23/49 (46.94%)) and Group 2 (from 0/27 (0%) to 10/25 (40%)), but negative seroconversion was observed in Group 4 (from 282/0 (100%) to 248/257 (96.50%)); while, all subjects in Group 3 remained seropositive at the end of monitoring. This evidence suggested for hybrid immunity, on which infection and vaccine simultaneously contributes to anti-RBD-SARS-CoV-2 IgG seroconversion. CONCLUSIONS A high seroprevalence of the IgG antibody against RBD-SARS-CoV-2 in the Sleman population was found to correlate with COVID-19 vaccination and as infection occurred, thus enhancing hybrid immunity. We also identified nonresponder and rapid antibody decaying individuals, that call for targeted vaccinations in addition to annual universal boosting.
Collapse
Affiliation(s)
- Jajah Fachiroh
- Department of Histology and Cell Biology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Septi Kurnia Lestari
- Sleman Health and Demographic Surveillance System, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Dewi Kartikawati Paramita
- Department of Histology and Cell Biology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Bagas Suryo Bintoro
- Department of Health Behavior, Environment and Social Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Center of Health Behavior and Promotion, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Fatwa Sari Tetra Dewi
- Sleman Health and Demographic Surveillance System, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Department of Health Behavior, Environment and Social Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Lutfan Lazuardi
- Department of Health Policy and Management, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Cici Permata Rusadi
- Sleman Health and Demographic Surveillance System, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Erti Nur Sagenah
- Sleman Health and Demographic Surveillance System, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Eggi Arguni
- Sleman Health and Demographic Surveillance System, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Department of Child Health, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Center for Tropical Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
18
|
Naffeti B, Ounissi Z, Srivastav AK, Stollenwerk N, Van-Dierdonck JB, Aguiar M. Modeling COVID-19 dynamics in the Basque Country: characterizing population immunity profile from 2020 to 2022. BMC Infect Dis 2025; 25:9. [PMID: 39748283 PMCID: PMC11697651 DOI: 10.1186/s12879-024-10342-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND COVID-19, caused by SARS-CoV-2, has spread globally, presenting a significant public health challenge. Vaccination has played a critical role in reducing severe disease and deaths. However, the waning of immunity after vaccination and the emergence of immune-escape variants require the continuation of vaccination efforts, including booster doses, to maintain population immunity. This study models the dynamics of COVID-19 in the Basque Country, Spain, aiming to characterize the population's immunity profile and assess its impact on the severity of outbreaks from 2020 to 2022. METHODS A SIR/DS model was developed to analyze the interplay of virus-specific and vaccine-induced immunity. The model includes three levels of immunity, with boosting effects from reinfection and/or vaccination. It was validated using empirical daily case data from the Basque Country. The model tracks shifts in immunity status and their effects on disease dynamics over time. RESULTS The COVID-19 epidemic in the Basque Country progressed through three distinct phases, each shaped by dynamic interactions between virus transmission, public health interventions, and vaccination efforts. The initial phase was marked by a rapid surge in cases, followed by a decline due to strict public health measures, with a seroprevalence of 1.3 % . In the intermediate phase, multiple smaller outbreaks emerged as restrictions were relaxed and new variants, such as Alpha and Delta, appeared. During this period, reinfection rates reached 20 % , and seroprevalence increased to 32 % . The final phase, dominated by the Omicron variant, saw a significant rise in cases driven by waning immunity and the variant's high transmissibility. Notably, 34 % of infections during this phase occurred in the naive population, with seroprevalence peaking at 43 % . Across all phases, the infection of naive and unvaccinated individuals contributed significantly to the severity of outbreaks, emphasizing the critical role of vaccination in mitigating disease impact. CONCLUSION The findings underscore the importance of continuous monitoring and adaptive public health strategies to mitigate the evolving epidemiological and immunological landscape of COVID-19. Dynamic interactions between immunity levels, reinfections, and vaccinations are critical in shaping outbreak severity and guiding evidence-based interventions.
Collapse
Affiliation(s)
- Bechir Naffeti
- Basque Center for Applied Mathematics, Bilbao, Bizkaia, Spain.
| | - Zeineb Ounissi
- Basque Center for Applied Mathematics, Bilbao, Bizkaia, Spain
| | | | | | | | - Maíra Aguiar
- Basque Center for Applied Mathematics, Bilbao, Bizkaia, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
19
|
Arévalo‐Herrera M, Rincón‐Orozco B, González‐Escobar JM, Herrera‐Arévalo SM, Carrasquilla‐Agudelo E, Serna‐Ortega PA, Quiceno‐García S, Palacio‐Muñoz N, Rosero‐López B, Mondol‐Miranda E, Freyle‐Roman I, Mendoza‐Landinez B, Mora‐Guevara E, Santos‐Barbosa JC, Bohórquez‐Martínez F, Bolaños‐Cristancho N, Jiménez‐Serna M, Nieto‐Rojas MA, Suarez‐Zamora D, Quintero‐Espinosa J, Londoño‐Trujillo D, Herrera‐ Valencia S. Longitudinal Follow-Up of the Specific Antibody Response to SARS-CoV-2 Vaccination in Colombia. J Med Virol 2025; 97:e70133. [PMID: 39817585 PMCID: PMC11737005 DOI: 10.1002/jmv.70133] [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: 05/22/2024] [Revised: 11/04/2024] [Accepted: 12/06/2024] [Indexed: 01/18/2025]
Abstract
A total of 5011 adult volunteers attending vaccination centers in different regions of Colombia were enrolled in a 1-year prospective observational cohort study to evaluate the immunogenicity and effectiveness of SARS-CoV-2-based vaccines as part of a National Vaccine Program established to contain the COVID-19 pandemic. Following informed consent, 5,011 participants underwent a sociodemographic survey and PCR testing to assess SARS-CoV-2 infection. Blood samples were collected, and serum fractions were obtained from a participant subsample (n = 3441) at six-time points to assess virus-specific IgG responses to the Spike protein, its Receptor Binding Domain, and the Nucleoprotein by ELISA. Additionally, antibody-neutralizing activity was evaluated using a cPass SARS-CoV-2 neutralization kit. Most participants (95.8%; n = 4802) received between one Ad26. COV2.S (Janssen vaccine) and four vaccine doses of BNT162b2 (Pfizer/BioNTech), AZD1222 (AstraZeneca), mRNA-1273 (Moderna), CoronaVac (Sinovac), with some receiving vaccine combinations; a small group, 4.2% (n = 209), remained unvaccinated. Throughout the study, only 8.76% (n = 439) of the participants tested positive for SARS-CoV-2 by PCR. Notably, all participants seroconverted for IgG antibodies, with high seropositivity rates for S (99.8%; n = 4795), RBD (99.7%; n = 1691), and N (92.7%; n = 3072) proteins. Moreover, significant (92%-97%) neutralizing activity was observed for all four SARS-CoV-2 circulating variants. This study highlights the importance of assessing the duration of the IgG response to SARS-CoV-2 elicited by vaccination and infection, and the antibody neutralizing activity as a potential surrogate marker of protection. These findings provide important insight for further strengthening the vaccination strategies to control COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Juliana Quintero‐Espinosa
- Fundación Santa Fe de BogotáSalud PoblacionalColombia
- Departamento de Medicina Interna, Sección de Infectología, Fundación Santa Fe de Bogotá
| | | | | |
Collapse
|
20
|
Gurry CE, Mwenda JM, Nardone A, Cohuet S, Worwui A, Valenciano M, Lewis HC, Wiysonge CS, Katsande R, Mukaro R, Braka F, Gueye AS, Balde T, Bergeri I, Impouma B. Establishing the African region monitoring vaccine effectiveness (AFRO-MoVE) network for respiratory pathogens. Vaccine 2024:126616. [PMID: 39743459 DOI: 10.1016/j.vaccine.2024.126616] [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: 10/24/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 01/04/2025]
Abstract
Population-level vaccination with newly developed vaccines to respond to the COVID-19 pandemic created a need to monitor vaccine effectiveness (VE) in the context of emerging SARS-CoV-2 variants and changing epidemiology. WHO and partners launched the African Region Monitoring Vaccine Effectiveness (AFRO-MoVE) Network in March 2021 to assess the performance of COVID-19 vaccines in real-world conditions in Africa. The Network aimed to facilitate and support comparable COVID-19 vaccine effectiveness studies in the African region, to provide a platform of scientific expertise and infrastructure, encourage the use of robust similar study designs to enable pooling to produce regional VE estimates and to build a sustainable network of hospitals, institutions, and experts to evaluate vaccines against pandemic and endemic respiratory pathogens. In the two years since its inception, the network has coordinated VE studies in the region and provided technical guidance and generic protocols employing robust methodologies. It brought together over 200 experts, representing 22 African countries and 55 organisations, and strengthened capacities by hosting ten webinars and six technical workshops. Of the 55 partners organisations, 25 based in 13 countries collaborated on implementing VE studies in the region. AFRO-MoVE supported study implementation in two phases, first targeting COVID-19 vaccination priority groups, then the general population. The network provides technical and financial support to nine studies, including three cohort studies in health workers and adults with comorbidities, and six test-negative design studies evaluating VE against symptomatic and severe disease. A data platform was established for pooled regional estimates. The AFRO-MoVE Network can form a sustainable platform to provide data for evidence informed decisions and timely VE monitoring for existing and new vaccines against respiratory pathogens and other diseases in the African region. Further development and consolidation of the network's activities can enable rapid response to future epidemics and pandemics.
Collapse
Affiliation(s)
- C E Gurry
- Epidemiology Department, Epiconcept, Paris, France.
| | - J M Mwenda
- World Health Organization Regional Office for Africa, Brazzaville, Congo
| | - A Nardone
- Epidemiology Department, Epiconcept, Paris, France
| | - S Cohuet
- Epidemiology Department, Epiconcept, Paris, France
| | - A Worwui
- World Health Organization Regional Office for Africa, Brazzaville, Congo
| | - M Valenciano
- Epidemiology Department, Epiconcept, Paris, France
| | - H C Lewis
- World Health Organization, Geneva, Switzerland
| | - C S Wiysonge
- World Health Organization Regional Office for Africa, Brazzaville, Congo
| | - R Katsande
- World Health Organization Regional Office for Africa, Brazzaville, Congo
| | - R Mukaro
- World Health Organization Regional Office for Africa, Brazzaville, Congo
| | - F Braka
- World Health Organization Regional Office for Africa, Brazzaville, Congo
| | - A S Gueye
- World Health Organization Regional Office for Africa, Brazzaville, Congo
| | - T Balde
- World Health Organization Regional Office for Africa, Brazzaville, Congo
| | - I Bergeri
- World Health Organization, Geneva, Switzerland
| | - B Impouma
- World Health Organization Regional Office for Africa, Brazzaville, Congo
| |
Collapse
|
21
|
Sergeeva MV, Vasilev K, Romanovskaya-Romanko E, Yolshin N, Pulkina A, Shamakova D, Shurygina AP, Muzhikyan A, Lioznov D, Stukova M. Mucosal Immunization with an Influenza Vector Carrying SARS-CoV-2 N Protein Protects Naïve Mice and Prevents Disease Enhancement in Seropositive Th2-Prone Mice. Vaccines (Basel) 2024; 13:15. [PMID: 39852794 PMCID: PMC11769390 DOI: 10.3390/vaccines13010015] [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: 12/05/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: Intranasal vaccination enhances protection against respiratory viruses by providing stimuli to the immune system at the primary site of infection, promoting a balanced and effective response. Influenza vectors with truncated NS1 are a promising vaccine approach that ensures a pronounced local CD8+ T-cellular immune response. Here, we describe the protective and immunomodulating properties of an influenza vector FluVec-N carrying the C-terminal fragment of the SARS-CoV-2 nucleoprotein within a truncated NS1 open reading frame. Methods: We generated several FluVec-N recombinant vectors by reverse genetics and confirmed the vector's genetic stability, antigen expression in vitro, attenuation, and immunogenicity in a mouse model. We tested the protective potential of FluVec-N intranasal immunization in naïve mice and seropositive Th2-prone mice, primed with aluminium-adjuvanted inactivated SARS-CoV-2. Immune response in immunized and challenged mice was analyzed through serological methods and flow cytometry. Results: Double intranasal immunization of naïve mice with FluVec-N reduced weight loss and viral load in the lungs following infection with the SARS-CoV-2 beta variant. Mice primed with alum-adjuvanted inactivated coronavirus experienced substantial early weight loss and eosinophilia in the lungs during infection, demonstrating signs of enhanced disease. A single intranasal boost immunization with FluVec-N prevented the disease enhancement in primed mice by modulating the local immune response. Protection was associated with the formation of specific IgA and the early activation of virus-specific effector and resident CD8+ lymphocytes in mouse lungs. Conclusions: Our study supports the potential of immunization with influenza vector vaccines to prevent respiratory diseases and associated immunopathology.
Collapse
Affiliation(s)
- Mariia V. Sergeeva
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia; (K.V.)
| | | | | | | | | | | | | | | | | | - Marina Stukova
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia; (K.V.)
| |
Collapse
|
22
|
Finkbeiner A, Khatib A, Upham N, Sterner B. A Systematic Review of the Distribution and Prevalence of Viruses Detected in the Peromyscus maniculatus Species Complex (Rodentia: Cricetidae). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.04.602117. [PMID: 39026800 PMCID: PMC11257420 DOI: 10.1101/2024.07.04.602117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The North American Deermouse, Peromyscus maniculatus, is one of the most widespread and abundant mammals on the continent. It is of public health interest as a known host of several viruses that are transmissible to humans and can cause illness, including the acute respiratory disease Hantavirus pulmonary syndrome (HPS). However, recent taxonomic studies indicate that P. maniculatus is a complex of multiple species, raising questions about how to identify and interpret three decades of hantavirus monitoring data. We conducted a systematic review investigating the prevalence and spatial distribution of viral taxa detected in wild populations allocated to P. maniculatus. From the 49 relevant studies published from 2000 to 2022, we extracted and analyzed spatial occurrence data to calculate weighted populational prevalences for hantaviruses. We found that detection efforts have been concentrated in the Western United States and Mexico with a focus on the spread of Sin Nombre virus (Orthohantavirus sinnombreense), the primary causative agent of HPS. There are significant gaps in the existing literature both geographically and in regard to the types of viruses being sampled. These results are significantly impacted by a recent taxonomic split of P. maniculatus into four species, and we were able to update 94% of hantavirus observations to reflect this change. Investigating the uncertain, and likely multiple, phylogenetic histories of these viral hosts should be a key emphasis of future modeling efforts.
Collapse
Affiliation(s)
| | - Ahmad Khatib
- School of Life Sciences, Arizona State University
| | - Nathan Upham
- School of Life Sciences, Arizona State University
| | | |
Collapse
|
23
|
Yamamoto M, Sakurai K, Takatani R, Hisada A, Mori C. Three-year seroprevalence of SARS-CoV-2 nucleocapsid protein antibody among children, parental awareness, and contributors of infection: a single-school cohort study in Chiba, Japan. J Epidemiol 2024; 35:278-286. [PMID: 39710421 PMCID: PMC12066190 DOI: 10.2188/jea.je20240284] [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: 03/21/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) in children is often asymptomatic, posing challenges in detecting infections. Additionally, factors contributing to infection remain poorly understood. This study aimed to investigate trends in anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleocapsid antibody seroprevalence, the relationship between seroprevalence and parental perception of child infection, and factors related to COVID-19 in children. METHODS In December 2020, 355 children aged 6-12 years in one elementary school were enrolled in the study. The anti-SARS-CoV-2 nucleocapsid antibody seroprevalence was assessed, and questionnaires were administered annually for three years. Parents' perceptions of infection and factors contributing to infection were examined. RESULTS The seroprevalence was 0.6%, 2.2%, and 60.9% in the first, second, and third years, respectively. The third-year seroprevalence among children reported as 'infected,' 'not tested but had symptoms,' and 'not infected' by parents was 97.3%, 83.3%, and 35.7%, respectively. Increased odds of seropositivity at the third-year measurement were observed in lower grades (adjusted odds ratio [aOR]=2.79 compared with higher grades) and in children more likely to play with others (aOR=3.97 for 'somewhat' and 2.84 for 'often,' compared with 'rarely'). No significant associations with seropositivity were found for sex, siblings, body mass index, serum 25-OH vitamin D3 concentration, or sleep duration. CONCLUSION The Omicron variant outbreak from the end of 2021 led to a sharp increase in seroprevalence among children, with many unaware of their infection. Frequent play with others may facilitate transmission in children. These data provide useful information for developing countermeasures against COVID-19 and other future pandemics.
Collapse
Affiliation(s)
- Midori Yamamoto
- Department of Sustainable Health Science, Center for Preventive Medical Sciences, Chiba University, Chiba, Japan
| | - Kenichi Sakurai
- Department of Nutrition and Metabolic Medicine, Center for Preventive Medical Sciences, Chiba University, Chiba, Japan
| | - Rieko Takatani
- Department of Clinical Medicine, Faculty of Education, Chiba University, Chiba, Japan
| | - Aya Hisada
- Department of Sustainable Health Science, Center for Preventive Medical Sciences, Chiba University, Chiba, Japan
| | - Chisato Mori
- Department of Sustainable Health Science, Center for Preventive Medical Sciences, Chiba University, Chiba, Japan
- Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
24
|
McNaughton CD, Austin PC, Li Z, Sivaswamy A, Fang J, Abdel-Qadir H, Udell JA, Wodchis WP, Lee DS, Mostarac I, Atzema CL. Higher Post-Acute Health Care Costs Following SARS-CoV-2 Infection Among Adults in Ontario, Canada. J Multidiscip Healthc 2024; 17:5749-5761. [PMID: 39659735 PMCID: PMC11628314 DOI: 10.2147/jmdh.s465154] [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: 04/09/2024] [Accepted: 09/06/2024] [Indexed: 12/12/2024] Open
Abstract
Purpose and Introduction Growing evidence suggests SARS-CoV-2 infection increases the risk of long term cardiovascular, neurological, and other effects. However, post-acute health care costs following SARS-CoV-2 infection are not known. Patients and Statistical Methods Beginning 56 days following SARS-CoV-2 polymerase chain reaction (PCR) testing, we compared person-specific total and component health care costs (2020 CAD$) for the first year of follow-up at the mean and 99th percentiles of health care costs for matched test-positive and test-negative adults in Ontario, Canada, between January 1, 2020, and March 31, 2021. Matching included demographics, baseline clinical characteristics, and two-week time blocks. Results For 531,182 people, mean person-specific total health care costs were $513.83 (95% CI $387.37-$638.40) higher for test-positive females and $459.10 (95% CI $304.60-$615.32) higher for test-positive males, which were driven by hospitalization, long-term care, and complex continuing care costs. At the 99th percentile of each subgroup, person-specific health care costs were $12,533.00 (95% CI $9008.50-$16,473.00) higher for test-positive females and $14,604.00 (95% CI $9565.50-$19,506.50) for test-positive males, driven by hospitalization, specialist (males), and homecare costs (females). Cancer costs were lower. Six-month and 1-year cost differences were similar. Conclusion Post-acute health care costs after a positive SARS-CoV-2 PCR test were significantly higher than matched test-negative individuals, and these increased costs persisted for at least one year. The largest increases health care costs came from hospitalizations, long-term care, complex continuing care, followed by outpatient specialists (for males) and homecare costs (for women). Given the magnitude of ongoing viral spread, policymakers, clinicians, and patients should be aware of higher post-acute health care costs following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Candace D McNaughton
- ICES (Formerly, the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
- Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, Toronto, ON, Canada
| | - Peter C Austin
- ICES (Formerly, the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
- Sunnybrook Research Institute, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, Toronto, ON, Canada
| | - Zhiyin Li
- ICES (Formerly, the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
| | - Atul Sivaswamy
- ICES (Formerly, the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
| | - Jiming Fang
- ICES (Formerly, the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
| | - Husam Abdel-Qadir
- ICES (Formerly, the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, Toronto, ON, Canada
- Division of Cardiology, Women’s College Hospital, Toronto, Ontario, Canada
| | - Jacob A Udell
- ICES (Formerly, the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
- Division of Cardiology, Women’s College Hospital, Toronto, Ontario, Canada
| | - Walter P Wodchis
- ICES (Formerly, the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, Toronto, ON, Canada
- Institute for Better Health, Trillium Health Partners, Mississauga, ON, Canada
| | - Douglas S Lee
- ICES (Formerly, the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, Toronto, ON, Canada
- Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada
| | | | - Clare L Atzema
- ICES (Formerly, the Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
- Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, Toronto, ON, Canada
| |
Collapse
|
25
|
Zeller I, Weiss A, Arnolds S, Schütte-Borkovec K, Arabi S, von dem Berge T, Casteels K, Hommel A, Kordonouri O, Larsson HE, Lundgren M, Rochtus A, Snape MD, Szypowka A, Vatish M, Winkler C, Bonifacio E, Ziegler AG. Infection episodes and islet autoantibodies in children at increased risk for type 1 diabetes before and during the COVID-19 pandemic. Infection 2024; 52:2465-2473. [PMID: 38874748 PMCID: PMC11621198 DOI: 10.1007/s15010-024-02312-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVES To determine the impact of the COVID-19 pandemic on the incidence rates of infection and islet autoimmunity in children at risk for type 1 diabetes. METHODS 1050 children aged 4 to 7 months with an elevated genetic risk for type 1 diabetes were recruited from Germany, Poland, Sweden, Belgium and the UK. Reported infection episodes and islet autoantibody development were monitored until age 40 months from February 2018 to February 2023. RESULTS The overall infection rate was 311 (95% Confidence Interval [CI], 304-318) per 100 person years. Infection rates differed by age, country, family history of type 1 diabetes, and period relative to the pandemic. Total infection rates were 321 per 100 person-years (95% CI 304-338) in the pre-pandemic period (until February 2020), 160 (95% CI 148-173) per 100 person-years in the first pandemic year (March 2020-February 2021; P < 0.001) and 337 (95% CI 315-363) per 100 person-years in subsequent years. Similar trends were observed for respiratory and gastrointestinal infections. Islet autoantibody incidence rates were 1.6 (95% CI 1.0-2.4) per 100 person-years in the pre-pandemic period, 1.2 (95% CI 0.8-1.9) per 100 person-years in the first pandemic year (P = 0.46), and 3.4 (95% CI 2.3-4.8) per 100 person-years in subsequent years (P = 0.005 vs. pre-pandemic year; P < 0.001 vs. first pandemic year). CONCLUSIONS The COVID-19 pandemic was associated with significantly altered infection patterns. Islet autoantibody incidence rates increased two-fold when infection rates returned to pre-pandemic levels.
Collapse
Affiliation(s)
- Ivo Zeller
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Heidemannstrasse 1, 80939, Munich, Germany
| | - Andreas Weiss
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Heidemannstrasse 1, 80939, Munich, Germany
| | - Stefanie Arnolds
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Heidemannstrasse 1, 80939, Munich, Germany
| | - Katharina Schütte-Borkovec
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Heidemannstrasse 1, 80939, Munich, Germany
| | - Sari Arabi
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- Department of Pediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | | | - Kristina Casteels
- Department of Pediatrics, University Hospitals Leuven, Louvain, Belgium
- Department of Development and Regeneration, KU Leuven, Louvain, Belgium
| | - Angela Hommel
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Olga Kordonouri
- Kinder- Und Jugendkrankenhaus AUF DER BULT, Hannover, Germany
| | - Helena Elding Larsson
- Unit for Pediatric Endocrinology, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Paediatrics, Skåne University Hospital, Malmö/Lund, Sweden
| | - Markus Lundgren
- Unit for Pediatric Endocrinology, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Pediatrics, Kristianstad Hospital, Kristianstad, Sweden
| | - Anne Rochtus
- Department of Pediatrics, University Hospitals Leuven, Louvain, Belgium
- Department of Development and Regeneration, KU Leuven, Louvain, Belgium
| | - Matthew D Snape
- Oxford Vaccine Group, University of Oxford Department of Paediatrics, NIHR Oxford Biomedical Research Centre, Oxford, UK
| | | | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health, Oxford, UK
| | - Christiane Winkler
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Heidemannstrasse 1, 80939, Munich, Germany
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Munich at University Hospital Carl Gustav Carus, Faculty of Medicine, TU, Dresden, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Heidemannstrasse 1, 80939, Munich, Germany.
- Forschergruppe Diabetes E.V. at Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany.
- Forschergruppe Diabetes, School of Medicine, Klinikum Rechts Der Isar, Technical University Munich, Munich, Germany.
| |
Collapse
|
26
|
Park WB, Hwang YH, Kwon KT, Noh JY, Park SH, Song JY, Choo EJ, Choi MJ, Choi JY, Heo JY, Choi WS. COVID-19 Vaccination Recommendations for 2024-2025 in Korea. Infect Chemother 2024; 56:453-460. [PMID: 39762924 PMCID: PMC11704866 DOI: 10.3947/ic.2024.0142] [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: 12/05/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
The Korean Society of Infectious Diseases has been regularly publishing guidelines for adult immunization since 2007. Following the release of coronavirus disease 2019 (COVID-19) vaccination recommendations in 2023, significant changes have occurred due to the emergence of new variant strains and the waning immunity from previous vaccinations. This article provides a comprehensive update as of November 2024, incorporating the latest evidence and guidelines. Focusing on the 2024-2025 season, this article reviews vaccines currently authorized in Korea and assesses their effectiveness against the predominant JN.1 lineage variants. The updated recommendations prioritize high-risk groups, including adults aged 65 and older, individuals with underlying medical conditions, residents of facilities vulnerable to infection, pregnant women, and healthcare workers, for vaccination with updated vaccines targeting the JN.1 strain. Additionally, COVID-19 vaccination is available for all individuals aged 6 months and older. For most adults, a single-dose strategy is emphasized, while tailored schedules may be recommended for immunocompromised individuals. This update aims to optimize vaccination strategies in Korea to ensure comprehensive protection for high-risk populations.
Collapse
Affiliation(s)
- Wan Beom Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| | - Young Hoon Hwang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Ki Tae Kwon
- Division of Infectious Diseases, Department of Internal Medicine, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Ji Yun Noh
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sun Hee Park
- Division of Infectious Diseases, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Joon Young Song
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Eun Ju Choo
- Division of Infectious Diseases, Department of Internal Medicine, College of Medicine, Soonchunhyang University of Korea, Bucheon, Korea
| | - Min Joo Choi
- Division of Infectious Diseases, International St Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon, Korea
| | - Jun Yong Choi
- Division of Infectious Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jung Yeon Heo
- Department of Infectious Diseases, Ajou University School of Medicine, Suwon, Korea
| | - Won Suk Choi
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
27
|
Berber E, Ross TM. Factors Predicting COVID-19 Vaccine Effectiveness and Longevity of Humoral Immune Responses. Vaccines (Basel) 2024; 12:1284. [PMID: 39591186 PMCID: PMC11598945 DOI: 10.3390/vaccines12111284] [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: 10/16/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, prompted global efforts to develop vaccines to control the disease. Various vaccines, including mRNA (BNT162b2, mRNA-1273), adenoviral vector (ChAdOx1, Ad26.COV2.S), and inactivated virus platforms (BBIBP-CorV, CoronaVac), elicit high-titer, protective antibodies against the virus, but long-term antibody durability and effectiveness vary. The objective of this study is to elucidate the factors that influence vaccine effectiveness (VE) and the longevity of humoral immune responses to COVID-19 vaccines through a review of the relevant literature, including clinical and real-world studies. Here, we discuss the humoral immune response to different COVID-19 vaccines and identify factors influencing VE and antibody longevity. Despite initial robust immune responses, vaccine-induced immunity wanes over time, particularly with the emergence of variants, such as Delta and Omicron, that exhibit immune escape mechanisms. Additionally, the durability of the humoral immune responses elicited by different vaccine platforms, along with the identification of essential determinants of long-term protection-like pre-existing immunity, booster doses, hybrid immunity, and demographic factors-are critical for protecting against severe COVID-19. Booster vaccinations substantially restore neutralizing antibody levels, especially against immune-evasive variants, while individuals with hybrid immunity have a more durable and potent immune response. Importantly, comorbidities such as diabetes, cardiovascular disease, chronic kidney disease, and cancer significantly reduce the magnitude and longevity of vaccine-induced protection. Immunocompromised individuals, particularly those undergoing chemotherapy and those with hematologic malignancies, have diminished humoral responses and benefit disproportionately from booster vaccinations. Age and sex also influence immune responses, with older adults experiencing accelerated antibody decline and females generally exhibiting stronger humoral responses compared to males. Understanding the variables affecting immune protection is crucial to improving vaccine strategies and predicting VE and protection against COVID-19.
Collapse
Affiliation(s)
- Engin Berber
- Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Ted M. Ross
- Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
- Florida Research and Innovation Center, Cleveland Clinic, Florida, FL 34986, USA
| |
Collapse
|
28
|
Carpp LN, Hyrien O, Fong Y, Benkeser D, Roels S, Stieh DJ, Van Dromme I, Van Roey GA, Kenny A, Huang Y, Carone M, McDermott AB, Houchens CR, Martins K, Jayashankar L, Castellino F, Amoa-Awua O, Basappa M, Flach B, Lin BC, Moore C, Naisan M, Naqvi M, Narpala S, O'Connell S, Mueller A, Serebryannyy L, Castro M, Wang J, Petropoulos CJ, Luedtke A, Lu Y, Yu C, Juraska M, Hejazi NS, Wolfe DN, Sadoff J, Gray GE, Grinsztejn B, Goepfert PA, Bekker LG, Gaur AH, Veloso VG, Randhawa AK, Andrasik MP, Hendriks J, Truyers C, Vandebosch A, Struyf F, Schuitemaker H, Douoguih M, Kublin JG, Corey L, Neuzil KM, Follmann D, Koup RA, Donis RO, Gilbert PB. Neutralizing antibody correlate of protection against severe-critical COVID-19 in the ENSEMBLE single-dose Ad26.COV2.S vaccine efficacy trial. Nat Commun 2024; 15:9785. [PMID: 39532861 PMCID: PMC11557889 DOI: 10.1038/s41467-024-53727-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Assessment of immune correlates of severe COVID-19 has been hampered by the low numbers of severe cases in COVID-19 vaccine efficacy (VE) trials. We assess neutralizing and binding antibody levels at 4 weeks post-Ad26.COV2.S vaccination as correlates of risk and of protection against severe-critical COVID-19 through 220 days post-vaccination in the ENSEMBLE trial (NCT04505722), constituting ~4.5 months longer follow-up than our previous correlates analysis and enabling inclusion of 42 severe-critical vaccine-breakthrough cases. Neutralizing antibody titer is a strong inverse correlate of severe-critical COVID-19, with estimated hazard ratio (HR) per 10-fold increase 0.35 (95% CI: 0.13, 0.90). In a multivariable model, HRs are 0.31 (0.11, 0.89) for neutralizing antibody titer and 1.22 (0.49, 3.02) for anti-Spike binding antibody concentration. VE against severe-critical COVID-19 rises with neutralizing antibody titer: 63.1% (95% CI: 40.0%, 77.3%) at unquantifiable [<4.8975 International Units (IU)50/ml], 85.2% (47.2%, 95.3%) at just-quantifiable (5.2 IU50/ml), and 95.1% (81.1%, 96.9%) at 90th percentile (30.2 IU50/ml). At the same titers, VE against moderate COVID-19 is 32.5% (11.8%, 48.4%), 33.9% (19.1%, 59.3%), and 60.7% (40.4%, 76.4%). Protection against moderate vs. severe disease may require higher antibody levels, and very low antibody levels and/or other immune responses may associate with protection against severe disease.
Collapse
Affiliation(s)
- Lindsay N Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - David Benkeser
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Sanne Roels
- Johnson & Johnson Innovative Medicine, Beerse, Belgium
| | - Daniel J Stieh
- Janssen Vaccines and Prevention, Leiden, the Netherlands
- Vaccine Company Inc., South San Francisco, CA, USA
| | | | | | - Avi Kenny
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
- Duke Global Health Institute, Duke University, Durham, NC, USA
| | - Ying Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Marco Carone
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Sanofi Vaccines R&D, Marcy l'étoile, France
| | | | - Karen Martins
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | - Flora Castellino
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Obrimpong Amoa-Awua
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Manjula Basappa
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Britta Flach
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bob C Lin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Moore
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mursal Naisan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Muhammed Naqvi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sandeep Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah O'Connell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Allen Mueller
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Leo Serebryannyy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mike Castro
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Alex Luedtke
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Yiwen Lu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Chenchen Yu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michal Juraska
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Nima S Hejazi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Daniel N Wolfe
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Jerald Sadoff
- Janssen Vaccines and Prevention, Leiden, the Netherlands
- Centivax, South San Francisco, CA, USA
| | - Glenda E Gray
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - Beatriz Grinsztejn
- Evandro Chagas National Institute of Infectious Diseases-Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Paul A Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Linda-Gail Bekker
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town, South Africa
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, Cape Town, South Africa
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Aditya H Gaur
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Valdilea G Veloso
- Evandro Chagas National Institute of Infectious Diseases-Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - April K Randhawa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michele P Andrasik
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jenny Hendriks
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | - Carla Truyers
- Johnson & Johnson Innovative Medicine, Beerse, Belgium
| | - An Vandebosch
- Johnson & Johnson Innovative Medicine, Beerse, Belgium
- argenx BV, Ghent, Belgium
| | - Frank Struyf
- Johnson & Johnson Innovative Medicine, Beerse, Belgium
- GSK, Wavre, Belgium
| | - Hanneke Schuitemaker
- Janssen Vaccines and Prevention, Leiden, the Netherlands
- Valneva, Saint-Herblain, France
| | - Macaya Douoguih
- Janssen Vaccines and Prevention, Leiden, the Netherlands
- Merck, Rahway, NJ, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
- Fogarty International Center, Bethesda, MD, USA
| | - Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ruben O Donis
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA.
| |
Collapse
|
29
|
Dezoumbe K, Djarma O, Wondeu ALD, Zakaria FA, Atturo S, Naïbeï N, Mennechet FJD, Campagna D, Boukar A, Moussa CA, Mahamat I, Armand N, Hamad MI, Honorine N, Frederic K, Moustapha AA, Daniel YM, Alim AM, Grene M, Suitombaye NY, Akouya A, Choua O, Mathieu H, Djimtoïbaye D, Colizzi V, Cappelli G, Rodrigue TDG, Abakar MF, Moussa AM. SARS-CoV-2 seroprevalence and associated factors among outpatient attendees at health facilities in different provinces in Chad. BMC Infect Dis 2024; 24:1271. [PMID: 39529023 PMCID: PMC11552332 DOI: 10.1186/s12879-024-10099-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Chad with 7,698 confirmed cases of infection and 194 deaths since the beginning of the COVID-19 pandemic, is one of the African countries with the lowest reported case numbers. However, this figure likely underestimates the true spread of the virus due to the low rate of diagnosis. The high rate of asymptomatic infections reflects the reality of SARS-CoV-2 transmission in Chad. In this study, we estimated the seroprevalence and identified factors associated with SARS-CoV-2 infection. METHODS A cross-sectional study was conducted between September 2022 and February 2023. A total of 1,290 plasma samples were collected from outpatient attendees at Health Facilities located in 11 provinces of Chad and tested by ELISA method, for the presence of IgG antibodies to SARS-CoV2 nucleocapsid (N) protein. KoboToolbox was used to gather data from the participants and data were analyzed using STATA 16. RESULTS The overall seroprevalence was 83.0% [95% CI = 81.6%-85.5%], with variations between provinces, ranging from 99.2% [95% CI = 94.0%-100%] in Moundou (Southern Chad) to 46.8% [95% CI = 36.0% -57.1%] in Biltine (Eastern Chad). Factors associated with the seroprevalence included military occupation (OR = 0.37 CI [0.80-1.77] p = 0.025) and age group between 55-64 years (OR = 0.33 CI [0.15-0.72] p = 0.005). While, other factors, such as gender and age were not significantly associated with seroprevalence. CONCLUSION Our results indicated that, the seroprevalence of COVID-19 in Chad is among the highest in Sub-Saharan Africa. These estimates could guide the response and public health policy decisions, enhancing the management of future outbreaks involving respiratory pathogens.
Collapse
Affiliation(s)
- Koutaya Dezoumbe
- Major Tropical Epidemics Laboratory "LAGET" of the , Good Samaritan University Hospital Centre, N'Djamena, Chad.
- Doctoral School of Advanced Sciences for Sustainable Developmentof the , Evangelical University of Cameroon, Bandjoun, Cameroon.
| | - Oumaima Djarma
- Department of Infectious Diseases, Le Bon Samaritain University, National University Hospital of Reference, N'Djamena, Chad
| | | | | | - Sabrina Atturo
- Italian Jesuit Movement and Action for Development Foundation (MAGIS), Rome, Italy
| | | | - Franck J D Mennechet
- Pathogenesis and Control of Chronic an Emerging Infections (PCCEI), University of Montpellier, French Blood Establishment (EFS), INSERM U1058, Montpellier, France
| | | | | | | | | | | | | | | | | | | | | | | | | | - Noubaramadji Yamti Suitombaye
- Major Tropical Epidemics Laboratory "LAGET" of the , Good Samaritan University Hospital Centre, N'Djamena, Chad
- Doctoral School of Advanced Sciences for Sustainable Developmentof the , Evangelical University of Cameroon, Bandjoun, Cameroon
| | - Amine Akouya
- Major Tropical Epidemics Laboratory "LAGET" of the , Good Samaritan University Hospital Centre, N'Djamena, Chad
- Doctoral School of Advanced Sciences for Sustainable Developmentof the , Evangelical University of Cameroon, Bandjoun, Cameroon
| | - Ouchemi Choua
- Faculty of Human Health Sciences, N'Djamena University, N'Djamena, Chad
- National University Hospital of Reference, N'Djamena, Chad
| | - Hota Mathieu
- Department of Laboratories, Ministry of Public Health, N'Djamena, Chad
| | - Djallaye Djimtoïbaye
- Major Tropical Epidemics Laboratory "LAGET" of the , Good Samaritan University Hospital Centre, N'Djamena, Chad
- Doctoral School of Advanced Sciences for Sustainable Developmentof the , Evangelical University of Cameroon, Bandjoun, Cameroon
| | - Vittorio Colizzi
- Evangelical University of Cameroon, Bandjoun, Cameroon
- Chantal Biya International Reference Center for Research on HIV/AIDS Prevention and Management (CIRCB), Yaoundé, Cameroon
- University of Rome "Tor Vergata", Rome, Italy
| | | | | | - Mahamat Fayiz Abakar
- Institut de Recherche en Elevage Pour Le Développement (IRED) Farcha, Chad, Farcha
| | - Ali Mahamat Moussa
- Faculty of Human Health Sciences, N'Djamena University, N'Djamena, Chad
- National University Hospital of Reference, N'Djamena, Chad
| |
Collapse
|
30
|
Boum Y, Matchim L, Guimsop DK, Buri BD, Bebell LM, Jaudel YSF, Njuwa FKG, Danirla DB, Youm E, Ntone R, Tchame CR, Tchiasso D, Essaka R, Eyong JB, Ngosso A, Nanda H, Fondze NR, Ndifon MN, Eteki L, Ghislain YFC, Messi BYE, Moustapha H, Hamdja M, Essomba RG, Mandeng N, Modeste TAK, Bisseck ACZK, Eyangoh SI, Njouom R, Okomo MC, Esso L, Emilienne E, Mballa GAE. High immunity and low mortality after Omicron and mass event in Cameroon despite low vaccination. J Public Health Afr 2024; 15:649. [PMID: 39649435 PMCID: PMC11622603 DOI: 10.4102/jphia.v15i1.649] [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: 06/01/2024] [Accepted: 09/07/2024] [Indexed: 12/10/2024] Open
Abstract
Background Little is known about the evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunity in African communities. Aim We evaluated changes in anti-SARS-CoV-2 antibodies, mortality and vaccination status in Cameroon between August 2021 and September 2022 to begin describing the evolution of the pandemic in Africa. Setting The study was conducted across Cameroon's 10 regional capitals, between 2021 and 2022 as the country hosted a mass gathering. Methods We conducted a cross-sectional population-based survey in 2022, including SARS-CoV-2 seroprevalence testing and retrospective mortality estimation using two-stage cluster sampling. We estimated and compared seroprevalence and crude mortality rates (CMR) to a survey conducted in 2021 using the same methodology. Results We performed serologic testing on 8400 individuals and collected mortality data from 22 314 individuals. Approximately 5% in each survey reported SARS-CoV-2-vaccination. Rapid diagnostic test-based seroprevalence increased from 11.2% (95% confidence interval [CI]: 10-12.5) to 59.8% (95% CI: 58.3-61.2) between 2021 and 2022, despite no increase in the proportion vaccinated. The CMR decreased from 0.17 to 0.06 deaths per 10 000 persons per day between 2021 and 2022. In 2022, no deaths were reportedly attributable to COVID-19 as compared to 17 deaths in 2021. Conclusion Over a 12-month period encompassing two waves of omicron variant SARS-CoV-2 and a mass gathering, SARS-CoV-2 seropositivity in Cameroon approached 60%, and deaths declined despite low vaccination coverage. Contribution This study challenges the assumption that high immunisation coverage is the sole determinant of epidemic control in the African context and encourages policymakers to increasingly rely on local research when designing response strategies for more effective outbreak management.
Collapse
Affiliation(s)
- Yap Boum
- Public Health Emergency Operation Center, Ministry of Public Health, Yaoundé, Cameroon
- Epicentre, Yaoundé, Cameroon
- Faculty of Biomedical Medicine and Science, University of Yaoundé I, Yaoundé, Cameroon
| | | | - Dominique K Guimsop
- Department for the Control of Disease, Epidemics and Pandemics, Ministry of Public Health, Yaoundé, Cameroon
- Western Africa Regional Coordination Center, Africa Centers for Disease Control, Abuja, Nigeria
| | | | - Lisa M Bebell
- Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, Massachusetts, United States of America
| | | | | | | | | | | | - Claudric Roosevelt Tchame
- Department for the Control of Disease, Epidemics and Pandemics, Ministry of Public Health, Yaoundé, Cameroon
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Nadia Mandeng
- Public Health Emergency Operation Center, Ministry of Public Health, Yaoundé, Cameroon
| | | | | | | | | | | | - Linda Esso
- Public Health Emergency Operation Center, Ministry of Public Health, Yaoundé, Cameroon
- Department for the Control of Disease, Epidemics and Pandemics, Yaoundé, Cameroon
| | - Epee Emilienne
- Public Health Emergency Operation Center, Ministry of Public Health, Yaoundé, Cameroon
- Department for the Control of Disease, Epidemics and Pandemics, Ministry of Public Health, Yaoundé, Cameroon
| | - Georges-Alain Etoundi Mballa
- Public Health Emergency Operation Center, Ministry of Public Health, Yaoundé, Cameroon
- Department for the Control of Disease, Epidemics and Pandemics, Ministry of Public Health, Yaoundé, Cameroon
| |
Collapse
|
31
|
Okada Y, Ueda M, Nishiura H. Reconstructing the age-structured case count of COVID-19 from sentinel surveillance data in Japan: A modeling study. Int J Infect Dis 2024; 148:107223. [PMID: 39209148 DOI: 10.1016/j.ijid.2024.107223] [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: 02/28/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVES To reconstruct age-structured case counts of COVID-19 using sentinel reporting, which replaced universal reporting of COVID-19 from May 2023 in Japan. METHODS Using COVID-19 sentinel data stratified by discrete age groups in selected prefectures and referring to universal case count data up to May 8, 2023, we fitted a statistical model to handle weekly growth rates as a function of age and time so as to convert sentinel data to case counts after cessation of universal reporting. RESULTS The age distribution of cases in sentinel reporting was significantly biased toward younger age groups compared to universal reporting. When comparing the epidemic size of the 9th wave (May 8 to September 18, 2023) to the 8th wave (October 3, 2022 to April 10, 2023), using the wave-on-wave ratio of total cumulative sentinel cases led to a significant underestimation of the wave-on-wave in Tokyo (0.975, vs 1.461 by universal reporting) and Okinawa (1.299, vs 1.472). The estimates of growth rates, scaling factors between universal and sentinel cases, and expected universal case count showed robustness to changes in the ending week of the data period. CONCLUSION Our model quantified COVID-19 dynamics, comparably to universal reporting that ended in May 2023, enabling detailed and up-to-date health burden analysis using sentinel reports. The cumulative incidence was greater than that suggested from sentinel data in Tokyo, Nara, and Okinawa. Per-population burdens among children were particularly high in Osaka and Nara, indicating a strong bias in sentinel reporting toward pediatric cases.
Collapse
Affiliation(s)
- Yuta Okada
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Minami Ueda
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | |
Collapse
|
32
|
Urwin E, Martin J, Sebire N, Harris A, Johnson J, Masood E, Milligan G, Mairs L, Chuter A, Ferguson M, Quinlan P, Jefferson E. A SARS-CoV-2 minimum data standard to support national serology reporting. Ann Clin Biochem 2024; 61:418-445. [PMID: 38806176 PMCID: PMC11528979 DOI: 10.1177/00045632241261274] [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] [Accepted: 05/13/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Healthcare laboratory systems produce and capture a vast array of information, yet do not always report all of this to the national infrastructure within the United Kingdom. The global COVID-19 pandemic brought about a much greater need for detailed healthcare data, one such instance being laboratory testing data. The reporting of qualitative laboratory test results (e.g. positive, negative or indeterminate) provides a basic understanding of levels of seropositivity. However, to better understand and interpret seropositivity, how it is determined and other factors that affect its calculation (i.e. levels of antibodies), quantitative laboratory test data are needed. METHOD 36 data attributes were collected from 3 NHS laboratories and 29 CO-CONNECT project partner organisations. These were assessed against the need for a minimum dataset to determine data attribute importance. An NHS laboratory feasibility study was undertaken to assess the minimum data standard, together with a literature review of national and international data standards and healthcare reports. RESULTS A COVID serology minimum data standard (CSMDS) comprising 12 data attributes was created and verified by 3 NHS laboratories to allow national granular reporting of COVID serology results. To support this, a standardised set of vocabulary terms was developed to represent laboratory analyser systems and laboratory information management systems. CONCLUSIONS This paper puts forward a minimum viable standard for COVID-19 serology data attributes to enhance its granularity and augment the national reporting of COVID-19 serology laboratory results, with implications for future pandemics.
Collapse
Affiliation(s)
- Esmond Urwin
- Digital Research Service, University of Nottingham, Nottingham, UK
| | - Joanne Martin
- Centre for Genomics and Child Health, Queen Mary University of London, London, UK
| | - Neil Sebire
- Institute of Child Health Population Policy and Practice, UCL Great Ormond Street Institute of Child Health, London, UK
| | | | - Jenny Johnson
- School of Medicine, University of Dundee, Dundee, UK
| | - Erum Masood
- School of Medicine, University of Dundee, Dundee, UK
| | | | | | - Antony Chuter
- Public and Patient Involvement Group, University of Nottingham, Nottingham, UK
| | | | - Philip Quinlan
- School of Medicine, University of Nottingham, Nottingham, UK
| | | |
Collapse
|
33
|
Ukwishaka J, Mela CF, Aseneh JBN, Ettaj M, Ilboudo D, Danwang C, Samadoulougou S, Kirakoya-Samadoulougou F. Seroprevalence of SARS-CoV-2 antibodies among healthy blood donors: a systematic review and meta-analysis. BMC Public Health 2024; 24:2925. [PMID: 39438911 PMCID: PMC11515703 DOI: 10.1186/s12889-024-20364-5] [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: 08/12/2023] [Accepted: 10/10/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION The development of a potent immune response and antibodies against SARS-CoV-2 is important for herd immunity. The serological response may be due to a previous infection or vaccination. Healthy blood donors could represent and provide information on the immune status of the general population. Therefore, we estimated the global and regional prevalence of SARS-CoV-2 antibodies among healthy blood donors. METHODS We conducted a systematic search of PubMed, Scopus, and ProQuest from December 2019 to January 2023. After critical appraisal and quality assessment, a qualitative synthesis of the identified relevant articles was performed. The random-effects model was used to estimate the pooled prevalence of SARS-CoV-2 antibodies. Funnel plots and Egger's test were used to assess publication bias. Sensitivity analysis was performed, and heterogeneity was quantified using I2 statistics. RESULTS A total of 70 peer-reviewed articles were selected that together included 2,454,192 blood donors. The global estimated pooled prevalence of SARS-CoV-2 antibodies among healthy blood donors was 10.3% (95% CI: 4.6 - 18.0%, n = 70). The highest seroprevalence was observed in Asia (17.7%), followed by Africa (16.1%). The seroprevalence in studies conducted before the introduction of the vaccine was 6.1%, whereas those of studies conducted after vaccines were available was 27.6%. High seroprevalence was observed in studies that measured antibodies against the S protein of the virus (15.2%), while lower (12.5%) in those that measured antibodies against the N protein. A high seroprevalence was observed in studies that only measured IgG antibodies (16.3%), and it was 5.9% in studies that measured total antibodies. CONCLUSION The prevalence of SARS-CoV-2 antibodies among healthy blood donors varies, potentially indicating geographical disparities in transmission and vaccination rates. To enhance community resilience, addressing these differences through inclusive health policies and adaptable public health measures is crucial.
Collapse
Affiliation(s)
- Joyeuse Ukwishaka
- Rwanda Biomedical Center, Maternal Child and Community Health Division, Kigali, Rwanda.
- Jhpiego, Kigali, Rwanda.
- Centre de Recherche en Epidémiologie, Biostatistique et Recherche Clinique, Ecole de Santé Publique, Université Libre de Bruxelles, Brussels, Belgium.
| | - Cyril Fotabong Mela
- Centre de Recherche en Epidémiologie, Biostatistique et Recherche Clinique, Ecole de Santé Publique, Université Libre de Bruxelles, Brussels, Belgium
| | - Jerry Brown Njoh Aseneh
- Centre de Recherche en Epidémiologie, Biostatistique et Recherche Clinique, Ecole de Santé Publique, Université Libre de Bruxelles, Brussels, Belgium
| | - Malak Ettaj
- Centre de Recherche en Epidémiologie, Biostatistique et Recherche Clinique, Ecole de Santé Publique, Université Libre de Bruxelles, Brussels, Belgium
- Department of Anthropology, University of California-Davis, Davis, CA, USA
| | - Dieudonné Ilboudo
- Centre de Recherche en Epidémiologie, Biostatistique et Recherche Clinique, Ecole de Santé Publique, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Sekou Samadoulougou
- Evaluation Platform on Obesity Prevention, Quebec Heart and Lung Institute, Quebec, QC, G1V 4G5, Canada
- Centre for Research on Planning and Development, Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Fati Kirakoya-Samadoulougou
- Centre de Recherche en Epidémiologie, Biostatistique et Recherche Clinique, Ecole de Santé Publique, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
34
|
Nycz E, Deng Y, Van Dyke ME, Bratcher A, Shi J, Iachan R, Berney K, Jones JM, Clarke KEN. School mask policies and SARS-CoV-2 seroprevalence among school-age children-United States, September to December 2021. Microbiol Spectr 2024; 12:e0069124. [PMID: 39194255 PMCID: PMC11449229 DOI: 10.1128/spectrum.00691-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/09/2024] [Indexed: 08/29/2024] Open
Abstract
During September to December 2021, school mask policies to mitigate SARS-CoV-2 transmission varied throughout the US. We compared infection-induced seroprevalence estimates and estimated seroconversion among children residing in areas with and without school mask requirements. We estimated infection-induced seroprevalence among children in three age groups (5-17, 5-11, and 12-17 years) in areas with and without school district mask requirements for two time points: September 1-30, 2021 and December 15, 2021 to January 14, 2022. Robust Poisson regression models estimated population seroconversion over the semester among initially seronegative children. Permutation tests assessed for significant differences in the estimated population seroconversion due to incident infections by school district mask policy. Residing in an area with no school mask requirement was associated with higher infection-induced seroprevalence among children aged 5-17 years (adjusted prevalence ratio [aPR] = 1.18, 95% confidence interval [CI]: 1.10, 1.26), and those aged 5-11 years (aPR) = 1.21, 95% CI: 1.10, 1.32) and those aged 12-17 years (aPR = 1.16, 95% CI: 1.07, 1.26), compared with areas requiring masks in school. Estimated population seroconversion during the semester was also significantly higher among children in districts without mask policies than those with school mask requirements among all age groups (5-17 years: 23.7% vs 18.1%, P < 0.001; 5-11 years: 6.4% vs 4.5%, P = 0.002;12-17 years: 27.2% vs 21.0%, P < 0.001). During the U.S. Fall 2021 semester, areas with school mask requirements had lower infection-induced seroprevalence and an estimated lower proportion of seroconversion due to incident infection among school-aged children compared with areas without school mask requirements; causality cannot necessarily be inferred from these associations. IMPORTANCE During the U.S. Fall 2021 school semester, the estimated proportion of previously uninfected school-aged children who experienced a first infection with SARS-CoV-2 was lower in areas where public school district policies required masks for all staff and students compared with areas where the school districts had no mask requirements. Because children are more likely than adults to experience asymptomatic or mild SARS-CoV-2 infections, the presence of infection-induced antibodies is a more accurate measure of infection history than clinical testing. The proportion of children with these antibodies (i.e., seroprevalence) can improve our understanding of SARS-CoV-2 by detecting more infections and eliminating potential bias due to local testing and reporting practices. Enhanced robustness of surveillance for respiratory infections in children, including records of mitigation policies in communities and schools, as well as seroprevalence data, would establish a better evidence base for policy decisions and response measures during future respiratory outbreaks.
Collapse
Affiliation(s)
- Elise Nycz
- COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Yangyang Deng
- ICF, Agency for Toxic Substances and Disease Registry, Atlanta, Georgia, USA
| | - Miriam E Van Dyke
- COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Anna Bratcher
- COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jianrong Shi
- COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Ronaldo Iachan
- ICF, Agency for Toxic Substances and Disease Registry, Atlanta, Georgia, USA
| | - Kevin Berney
- Geospatial Research, Analysis, and Services Program (GRASP), Agency for Toxic Substances and Disease Registry, Atlanta, Georgia, USA
| | - Jefferson M Jones
- COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Kristie E N Clarke
- COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
35
|
Cohen AA, Keeffe JR, Schiepers A, Dross SE, Greaney AJ, Rorick AV, Gao H, Gnanapragasam PNP, Fan C, West AP, Ramsingh AI, Erasmus JH, Pata JD, Muramatsu H, Pardi N, Lin PJC, Baxter S, Cruz R, Quintanar-Audelo M, Robb E, Serrano-Amatriain C, Magneschi L, Fotheringham IG, Fuller DH, Victora GD, Bjorkman PJ. Mosaic sarbecovirus nanoparticles elicit cross-reactive responses in pre-vaccinated animals. Cell 2024; 187:5554-5571.e19. [PMID: 39197450 PMCID: PMC11460329 DOI: 10.1016/j.cell.2024.07.052] [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: 01/17/2024] [Revised: 06/15/2024] [Accepted: 07/27/2024] [Indexed: 09/01/2024]
Abstract
Immunization with mosaic-8b (nanoparticles presenting 8 SARS-like betacoronavirus [sarbecovirus] receptor-binding domains [RBDs]) elicits more broadly cross-reactive antibodies than homotypic SARS-CoV-2 RBD-only nanoparticles and protects against sarbecoviruses. To investigate original antigenic sin (OAS) effects on mosaic-8b efficacy, we evaluated the effects of prior COVID-19 vaccinations in non-human primates and mice on anti-sarbecovirus responses elicited by mosaic-8b, admix-8b (8 homotypics), or homotypic SARS-CoV-2 immunizations, finding the greatest cross-reactivity for mosaic-8b. As demonstrated by molecular fate mapping, in which antibodies from specific cohorts of B cells are differentially detected, B cells primed by WA1 spike mRNA-LNP dominated antibody responses after RBD-nanoparticle boosting. While mosaic-8b- and homotypic-nanoparticles boosted cross-reactive antibodies, de novo antibodies were predominantly induced by mosaic-8b, and these were specific for variant RBDs with increased identity to RBDs on mosaic-8b. These results inform OAS mechanisms and support using mosaic-8b to protect COVID-19-vaccinated/infected humans against as-yet-unknown SARS-CoV-2 variants and animal sarbecoviruses with human spillover potential.
Collapse
Affiliation(s)
- Alexander A Cohen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jennifer R Keeffe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ariën Schiepers
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY 10065, USA
| | - Sandra E Dross
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA; National Primate Research Center, Seattle, WA 98121, USA
| | - Allison J Greaney
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Annie V Rorick
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Han Gao
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | - Chengcheng Fan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Anthony P West
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | | | - Janice D Pata
- Wadsworth Center, New York State Department of Health and Department of Biomedical Sciences, University at Albany, Albany, NY 12201, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Scott Baxter
- Ingenza Ltd., Roslin Innovation Centre, Charnock Bradley Building, Roslin EH25 9RG, UK
| | - Rita Cruz
- Ingenza Ltd., Roslin Innovation Centre, Charnock Bradley Building, Roslin EH25 9RG, UK
| | | | - Ellis Robb
- Ingenza Ltd., Roslin Innovation Centre, Charnock Bradley Building, Roslin EH25 9RG, UK
| | | | - Leonardo Magneschi
- Ingenza Ltd., Roslin Innovation Centre, Charnock Bradley Building, Roslin EH25 9RG, UK
| | - Ian G Fotheringham
- Ingenza Ltd., Roslin Innovation Centre, Charnock Bradley Building, Roslin EH25 9RG, UK
| | - Deborah H Fuller
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA; National Primate Research Center, Seattle, WA 98121, USA
| | - Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY 10065, USA
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
36
|
Sun K, Bhiman JN, Tempia S, Kleynhans J, Madzorera VS, Mkhize Q, Kaldine H, McMorrow ML, Wolter N, Moyes J, Carrim M, Martinson NA, Kahn K, Lebina L, du Toit JD, Mkhencele T, von Gottberg A, Viboud C, Moore PL, Cohen C. SARS-CoV-2 correlates of protection from infection against variants of concern. Nat Med 2024; 30:2805-2812. [PMID: 39060660 PMCID: PMC11533127 DOI: 10.1038/s41591-024-03131-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/11/2024] [Indexed: 07/28/2024]
Abstract
Serum neutralizing antibodies (nAbs) induced by vaccination have been linked to protection against symptomatic and severe coronavirus disease 2019. However, much less is known about the efficacy of nAbs in preventing the acquisition of infection, especially in the context of natural immunity and against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immune-escape variants. Here we conducted mediation analysis to assess serum nAbs induced by prior SARS-CoV-2 infections as potential correlates of protection against Delta and Omicron infections, in rural and urban household cohorts in South Africa. We find that, in the Delta wave, D614G nAbs mediate 37% (95% confidence interval: 34-40%) of the total protection against infection conferred by prior exposure to SARS-CoV-2, and that protection decreases with waning immunity. In contrast, Omicron BA.1 nAbs mediate 11% (95% confidence interval: 9-12%) of the total protection against Omicron BA.1 or BA.2 infections, due to Omicron's neutralization escape. These findings underscore that correlates of protection mediated through nAbs are variant specific, and that boosting of nAbs against circulating variants might restore or confer immune protection lost due to nAb waning and/or immune escape. However, the majority of immune protection against SARS-CoV-2 conferred by natural infection cannot be fully explained by serum nAbs alone. Measuring these and other immune markers including T cell responses, both in the serum and in other compartments such as the nasal mucosa, may be required to comprehensively understand and predict immune protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Kaiyuan Sun
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda, MD, USA.
| | - Jinal N Bhiman
- SAMRC Antibody Immunity Research Unit, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Stefano Tempia
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jackie Kleynhans
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Vimbai Sharon Madzorera
- SAMRC Antibody Immunity Research Unit, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Qiniso Mkhize
- SAMRC Antibody Immunity Research Unit, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Haajira Kaldine
- SAMRC Antibody Immunity Research Unit, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Meredith L McMorrow
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Nicole Wolter
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jocelyn Moyes
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Maimuna Carrim
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Neil A Martinson
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
- Johns Hopkins University Center for TB Research, Baltimore, MD, USA
| | - Kathleen Kahn
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Limakatso Lebina
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Jacques D du Toit
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Thulisa Mkhencele
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Anne von Gottberg
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Cécile Viboud
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda, MD, USA
| | - Penny L Moore
- SAMRC Antibody Immunity Research Unit, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Cheryl Cohen
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
37
|
Perez-Guzman PN, Chanda SL, Schaap A, Shanaube K, Baguelin M, Nyangu ST, Kanyanga MK, Walker P, Ayles H, Chilengi R, Verity R, Hauck K, Knock ES, Cori A. Pandemic burden in low-income settings and impact of limited and delayed interventions: A granular modelling analysis of COVID-19 in Kabwe, Zambia. Int J Infect Dis 2024; 147:107182. [PMID: 39067669 DOI: 10.1016/j.ijid.2024.107182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/04/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024] Open
Abstract
OBJECTIVES Pandemic response in low-income countries (LICs) or settings often suffers from scarce epidemic surveillance and constrained mitigation capacity. The drivers of pandemic burden in such settings, and the impact of limited and delayed interventions remain poorly understood. METHODS We analysed COVID-19 seroprevalence and all-cause excess deaths data from the peri-urban district of Kabwe, Zambia between March 2020 and September 2021 with a novel mathematical model. Data encompassed three consecutive waves caused by the wild-type, Beta and Delta variants. RESULTS Across all three waves, we estimated a high cumulative attack rate, with 78% (95% credible interval [CrI] 71-85) of the population infected, and a high all-cause excess mortality, at 402 (95% CrI 277-473) deaths per 100,000 people. Ambitiously improving health care to a capacity similar to that in high-income settings could have averted up to 46% (95% CrI 41-53) of accrued excess deaths, if implemented from June 2020 onward. An early and accelerated vaccination rollout could have achieved the highest reductions in deaths. Had vaccination started as in some high-income settings in December 2020 and with the same daily capacity (doses per 100 population), up to 68% (95% CrI 64-71) of accrued excess deaths could have been averted. Slower rollouts would have still averted 62% (95% CrI 58-68), 54% (95% CrI 49-61) or 26% (95% CrI 20-38) of excess deaths if matching the average vaccination capacity of upper-middle-, lower-middle- or LICs, respectively. CONCLUSIONS Robust quantitative analyses of pandemic data are of pressing need to inform future global pandemic preparedness commitments.
Collapse
Affiliation(s)
- Pablo N Perez-Guzman
- Imperial College London, Medical Research Council Centre for Global Infectious Disease Analysis, and Abdul Latif Jameel Institute for Disease and Emergency Analytics, School of Public Health, London, UK.
| | | | - Albertus Schaap
- Zambart, Lusaka, Zambia; London School of Hygiene & Tropical Medicine, Faculty of Infectious and Tropical Diseases, London, UK
| | | | - Marc Baguelin
- Imperial College London, Medical Research Council Centre for Global Infectious Disease Analysis, and Abdul Latif Jameel Institute for Disease and Emergency Analytics, School of Public Health, London, UK; National Institute for Health and Care Research, Health Protection Research Unit in Modelling and Health Economics, London, UK; London School of Hygiene & Tropical Medicine, Centre for Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, London, UK
| | | | | | - Patrick Walker
- Imperial College London, Medical Research Council Centre for Global Infectious Disease Analysis, and Abdul Latif Jameel Institute for Disease and Emergency Analytics, School of Public Health, London, UK
| | - Helen Ayles
- Zambart, Lusaka, Zambia; London School of Hygiene & Tropical Medicine, Faculty of Infectious and Tropical Diseases, London, UK
| | - Roma Chilengi
- Zambia National Public Health Institute, Lusaka, Zambia
| | - Robert Verity
- Imperial College London, Medical Research Council Centre for Global Infectious Disease Analysis, and Abdul Latif Jameel Institute for Disease and Emergency Analytics, School of Public Health, London, UK
| | - Katharina Hauck
- Imperial College London, Medical Research Council Centre for Global Infectious Disease Analysis, and Abdul Latif Jameel Institute for Disease and Emergency Analytics, School of Public Health, London, UK
| | - Edward S Knock
- Imperial College London, Medical Research Council Centre for Global Infectious Disease Analysis, and Abdul Latif Jameel Institute for Disease and Emergency Analytics, School of Public Health, London, UK
| | - Anne Cori
- Imperial College London, Medical Research Council Centre for Global Infectious Disease Analysis, and Abdul Latif Jameel Institute for Disease and Emergency Analytics, School of Public Health, London, UK
| |
Collapse
|
38
|
Volkman HR, Nguyen JL, Jodar L, McLaughlin JM. A Letter to the Editor Regarding 'Comparative Effectiveness of mRNA-1273 and BNT162b2 COVID-19 Vaccines Among Older Adults: Systematic Literature Review and Meta-analysis Using the GRADE Framework'. Infect Dis Ther 2024; 13:2203-2206. [PMID: 39180645 PMCID: PMC11416434 DOI: 10.1007/s40121-024-01019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/02/2024] [Indexed: 08/26/2024] Open
Affiliation(s)
| | | | - Luis Jodar
- Pfizer Inc., 66 Hudson Blvd. E, New York, NY, 10001, USA
| | | |
Collapse
|
39
|
Owusu-Boaitey N, Böttcher L, He D, Erkhembayar R, Yang L, Kim DH, Barchuk A, Gorski DH, Howard J. Impact of cross-reactivity and herd immunity on SARS-CoV-2 pandemic severity. Infect Dis (Lond) 2024; 56:897-902. [PMID: 39133617 DOI: 10.1080/23744235.2024.2388222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 07/31/2024] [Indexed: 10/01/2024] Open
Abstract
Public health systems reported low mortality from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in East Asia, in low-income countries, and for children during the first year of the SARS-CoV-2 pandemic. These reports led commentators to suggest that cross-reactive immunity from prior exposure to other pathogens reduced fatality risk. Resolution of initial infection waves also contributed to speculation that herd immunity prevented further waves prior to vaccination. Serology instead implied that immunity was too limited to achieve herd immunity and that there was little impact from cross-reactive protection. Paediatric deaths exceeded those from influenza, with higher age-specific fatality risk in lower-income nations and similar fatality risk in East Asia compared with demographically similar regions. Neither pre-outbreak exposure to related pathogens nor immunity induced by initial infection waves are necessarily a reliable response to future pathogen outbreaks. Preparedness for future pathogen outbreaks should instead focus on strategies such as voluntary behavioural changes, nonpharmaceutical interventions, and vaccination.
Collapse
Affiliation(s)
- Nana Owusu-Boaitey
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - Lucas Böttcher
- Department of Computational Science and Philosophy, Frankfurt School of Finance and Management, Frankfurt a. M, Germany
- Laboratory for Systems Medicine, Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Daihai He
- Department of Applied Mathematics, Hong Kong Polytechnic University, Hong Kong, China
| | - Ryenchindorj Erkhembayar
- International Cyber Education Center, Graduate School, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Lin Yang
- School of Nursing, Hong Kong Polytechnic University, Hong Kong, China
| | - Dong-Hyun Kim
- Department of Social and Preventive Medicine, Hallym University College of Medicine, Chuncheon, South Korea
| | - Anton Barchuk
- Institute for Interdisciplinary Health Research, European University at St. Petersburg, St. Petersburg, Russia
- Program for Public Health Sciences, ITMO University, St. Petersburg, Russia
| | - David H Gorski
- Departments of Surgery and Oncology, Wayne State University, Detroit, Michigan, USA
| | - Jonathan Howard
- Department of Neurology, NYU Langone Health, New York, New York, USA
| |
Collapse
|
40
|
Hannawi S, Abuquta A, Eldin LS, Hassan A, Alamadi A, Gao C, Baidoo AAH, Yang X, Su H, Zhang J, Xie L. Immunogenicity and Safety of Omicron-Containing Multivalent COVID-19 Vaccines in Unvaccinated and Previously Vaccinated Adults. Vaccines (Basel) 2024; 12:1109. [PMID: 39460276 PMCID: PMC11510771 DOI: 10.3390/vaccines12101109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
The SARS-CoV-2 evolution trajectory remains uncertain, and the antigenic characteristics of future variants are highly unpredictable. We report the immunogenicity and safety of multivalent COVID-19 vaccines, SCTV01E and SCTV01E-1, against Omicron BA.5. This phase 2 trial randomized 400 adults into two cohorts, 160 unvaccinated (3 doses) and 240 previously vaccinated (2 doses) individuals to receive 30 µg SCTV01E-1 or 30 µg SCTV01E (1:1) between 4 November and 28 November 2022. Among the unvaccinated cohort, day 42 geometric mean fold rises (GMFRs) of neutralizing antibodies (nAb) against Omicron BA.5 were reported to be 12.8× and 20.5× over day 0 for SCTV01E-1 and SCTV01E, respectively. On day 178, both vaccines increased geometric mean titers (GMTs) of nAb against BA.5 following the booster dose compared to pre-booster levels on D150. Similar frequencies of solicited [6.2% (5/81) and 7.6% (6/79)] and unsolicited [11.1% (9/81) and 10.1% (8/79)] adverse events (AEs) were reported in SCTV01E-1 and SCTV01E groups, respectively. Grade 3 or more AEs were < 2% in both vaccine groups [SCTV01E-1: 1.2% (1/81), SCTV01E: 1.3% (1/79)]. In the previously vaccinated cohort, similar GMFRs were reported on day 28 (SCTV01E-1: 9.4× and SCTV01E: 8.7×) over baseline (D0). On day 148, both vaccines showed increased nAb levels with similar GMFRs over D120. Comparable incidences of solicited [13.2% (16/121) and 10.9% (13/119)] and unsolicited [17.4% (21/121) and 10.9% (13/119)] AEs were reported in SCTV01E-1 and SCTV01E groups, respectively. Numerically identical ≥ grade 3 AEs [SCTV01E-1: 1.7% (2/121) and SCTV01E: 1.7% (2/119)] were reported. This trial demonstrates the effectiveness of updated multivalent vaccines with acceptable safety profiles.
Collapse
Affiliation(s)
- Suad Hannawi
- Internal Medicine Department, Al Kuwait-Dubai (ALBaraha) Hospital, Dubai 00000, United Arab Emirates; (S.H.); (A.H.)
| | - Alaa Abuquta
- Accident and Emergency Department, Al Kuwait-Dubai (ALBaraha) Hospital, Dubai 00000, United Arab Emirates;
| | - Linda Saf Eldin
- General Surgery Department, Al Kuwait-Dubai (ALBaraha) Hospital, Dubai 00000, United Arab Emirates;
| | - Aala Hassan
- Internal Medicine Department, Al Kuwait-Dubai (ALBaraha) Hospital, Dubai 00000, United Arab Emirates; (S.H.); (A.H.)
| | - Ahmad Alamadi
- Ear, Nose and Throat Department (ENT), Al Kuwait-Dubai (ALBaraha) Hospital, Dubai 00000, United Arab Emirates;
| | - Cuige Gao
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (C.G.); (A.A.H.B.); (X.Y.); (H.S.); (J.Z.)
| | - Adam Abdul Hakeem Baidoo
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (C.G.); (A.A.H.B.); (X.Y.); (H.S.); (J.Z.)
| | - Xinjie Yang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (C.G.); (A.A.H.B.); (X.Y.); (H.S.); (J.Z.)
| | - Huo Su
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (C.G.); (A.A.H.B.); (X.Y.); (H.S.); (J.Z.)
| | - Jinxiu Zhang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (C.G.); (A.A.H.B.); (X.Y.); (H.S.); (J.Z.)
| | - Liangzhi Xie
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (C.G.); (A.A.H.B.); (X.Y.); (H.S.); (J.Z.)
- Cell Culture Engineering Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
41
|
Zhang B, Fong Y, Fintzi J, Chu E, Janes HE, Kenny A, Carone M, Benkeser D, van der Laan LWP, Deng W, Zhou H, Wang X, Lu Y, Yu C, Borate B, Chen H, Reeder I, Carpp LN, Houchens CR, Martins K, Jayashankar L, Huynh C, Fichtenbaum CJ, Kalams S, Gay CL, Andrasik MP, Kublin JG, Corey L, Neuzil KM, Priddy F, Das R, Girard B, El Sahly HM, Baden LR, Jones T, Donis RO, Koup RA, Gilbert PB, Follmann D. Omicron COVID-19 immune correlates analysis of a third dose of mRNA-1273 in the COVE trial. Nat Commun 2024; 15:7954. [PMID: 39261482 PMCID: PMC11390939 DOI: 10.1038/s41467-024-52348-9] [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: 02/26/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024] Open
Abstract
In the phase 3 Coronavirus Efficacy (COVE) trial (NCT04470427), post-dose two Ancestral Spike-specific binding (bAb) and neutralizing (nAb) antibodies were shown to be correlates of risk (CoR) and of protection against Ancestral-lineage COVID-19 in SARS-CoV-2 naive participants. In the SARS-CoV-2 Omicron era, Omicron subvariants with varying degrees of immune escape now dominate, seropositivity rates are high, and booster doses are administered, raising questions on whether and how these developments affect the bAb and nAb correlates. To address these questions, we assess post-boost BA.1 Spike-specific bAbs and nAbs as CoRs and as correlates of booster efficacy in COVE. For naive individuals, bAbs and nAbs inversely correlate with Omicron COVID-19: hazard ratios (HR) per 10-fold marker increase (95% confidence interval) are 0.16 (0.03, 0.79) and 0.31 (0.10, 0.96), respectively. In non-naive individuals the analogous results are similar: 0.15 (0.04, 0.63) and 0.28 (0.07, 1.08). For naive individuals, three vs two-dose booster efficacy correlates with predicted nAb titer at exposure, with estimates -8% (-126%, 48%), 50% (25%, 67%), and 74% (49%, 87%), at 56, 251, and 891 Arbitrary Units/ml. These results support the continued use of antibody as a surrogate endpoint.
Collapse
Affiliation(s)
- Bo Zhang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jonathan Fintzi
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Eric Chu
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Holly E Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Avi Kenny
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Marco Carone
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - David Benkeser
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | | | | | | | | | - Yiwen Lu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Chenchen Yu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Bhavesh Borate
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Haiyan Chen
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Isabel Reeder
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Lindsay N Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Karen Martins
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | - Chuong Huynh
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Carl J Fichtenbaum
- Division of Infectious Diseases, Department of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Spyros Kalams
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cynthia L Gay
- Department of Medicine, Division of Infectious Diseases, UNC HIV Cure Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Michele P Andrasik
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
- Fogarty International Center, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - Hana M El Sahly
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Thomas Jones
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Ruben O Donis
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
42
|
Osman A, Aimone A, Ansumana R, Bogoch I, Gelband H, Colwill K, Gingras AC, Langlois MA, Carshon-Marsh R, Swaray IB, Jambai A, Vandi M, Vandi A, Massaquoi M, Assalif A, Birnboim HC, Brown PE, Nagelkerke N, Jha P. High SARS-CoV-2 seroincidence but low excess COVID mortality in Sierra Leone in 2020-2022. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003411. [PMID: 39255307 PMCID: PMC11386415 DOI: 10.1371/journal.pgph.0003411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/31/2024] [Indexed: 09/12/2024]
Abstract
While SARS-CoV-2 infection appears to have spread widely throughout Africa, documentation of associated mortality is limited. We implemented a representative serosurvey in one city of Sierra Leone in Western Africa, paired with nationally representative mortality and selected death registration data. Cumulative seroincidence using high quality SARS-CoV-2 serological assays was 69% by July 2021, rising to 84% by April 2022, mostly preceding SARS-CoV-2 vaccination. About half of infections showed evidence of neutralizing antibodies. However, excess death rates were low, and were concentrated at older ages. During the peak weeks of viral activity, excess mortality rates were 22% for individuals aged 30-69 years and 70% for those over 70. Based on electronic verbal autopsy with dual independent physician assignment of causes, excess deaths during viral peaks from respiratory infections were notable. Excess deaths differed little across specific causes that, a priori, are associated with COVID, and the pattern was consistent among adults with or without chronic disease risk factors. The overall 6% excess of deaths at ages ≥30 from 2020-2022 in Sierra Leone is markedly lower than reported from South Africa, India, and Latin America. Thus, while SARS-CoV-2 infection was widespread, our study highlights as yet unidentified mechanisms of heterogeneity in susceptibility to severe disease in parts of Africa.
Collapse
Affiliation(s)
- Ahmed Osman
- Centre for Global Health Research, Unity Health Toronto and Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- School of Community Health Sciences Njala University, Bo, Sierra Leone
| | - Ashley Aimone
- Centre for Global Health Research, Unity Health Toronto and Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Rashid Ansumana
- School of Community Health Sciences Njala University, Bo, Sierra Leone
| | - Isaac Bogoch
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hellen Gelband
- Centre for Global Health Research, Unity Health Toronto and Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Ibrahim Bob Swaray
- Centre for Global Health Research, Unity Health Toronto and Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Amara Jambai
- Ministry of Health, Government of Sierra Leone, Freetown, Sierra Leone
| | - Mohamed Vandi
- Ministry of Health, Government of Sierra Leone, Freetown, Sierra Leone
| | - Alimatu Vandi
- School of Community Health Sciences Njala University, Bo, Sierra Leone
| | | | - Anteneh Assalif
- School of Community Health Sciences Njala University, Bo, Sierra Leone
| | | | - Patrick E. Brown
- Centre for Global Health Research, Unity Health Toronto and Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Nico Nagelkerke
- Centre for Global Health Research, Unity Health Toronto and Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Prabhat Jha
- Centre for Global Health Research, Unity Health Toronto and Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Ramadani N, Hoxha‐Gashi S, Gexha‐Bunjaku D, Kalaveshi A, Jakupi X, Humolli I, Vaughan A, Pebody R, Kacaniku‐Gunga P, Jashari V. Prevalence of SARS-CoV-2 Antibodies in Kosovo-Wide Population-Based Seroepidemiological Study. Influenza Other Respir Viruses 2024; 18:e70004. [PMID: 39225019 PMCID: PMC11369638 DOI: 10.1111/irv.70004] [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: 02/06/2024] [Revised: 07/28/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Seroprevalence studies have proven to be an important tool in tracking the progression of the coronavirus disease 2019 (COVID-19) pandemic. The aim of this study was to measure the seroprevalence of antibodies to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the general population of Kosovo by gender, age group and region and among asymptomatic people. METHOD The Institute of Public Health of Kosovo conducted a cross-sectional population-based survey, aligned with the protocols of the WHO Unity Studies, from the beginning of May to the end of June 2021. RESULTS The survey covered a total of 2204 people with a response rate of 91.8% (41.9% [923] males and 51.2% [1281] females). In May to June 2021, the prevalence of antibodies in the overall population (IgG antibodies ≥ 1.1) was 37.0%. Seroprevalence was 34.4% in men and 38.9% in women (p < 0.05), with the highest percentage (48.7%) found in the 60-69 years' age group. The overall prevalence of acute IgM antibodies (IgM ≥ 1.1) was 1% (95% CI: 0.7%-1.5%), with no significant difference between genders and the highest prevalence among participants of 60-69 years of age (1.6%; 95% CI: 0.7%-3.6%). CONCLUSION A high prevalence of antibodies against SARS-CoV-2 was found in Kosovo before the start of the vaccination campaign. However, the results of the survey suggested that, by the end of June 2021, a desirable level of protection from the SARS-CoV-2 virus had not been reached.
Collapse
Affiliation(s)
- Naser Ramadani
- Faculty of MedicineUniversity of Pristina “Hasan Pristina”PristinaKosovo
- National Institute of Public HealthPristinaKosovo
| | - Sanije Hoxha‐Gashi
- Faculty of MedicineUniversity of Pristina “Hasan Pristina”PristinaKosovo
- National Institute of Public HealthPristinaKosovo
| | | | - Arijana Kalaveshi
- Faculty of MedicineUniversity of Pristina “Hasan Pristina”PristinaKosovo
- National Institute of Public HealthPristinaKosovo
| | - Xhevat Jakupi
- Faculty of MedicineUniversity of Pristina “Hasan Pristina”PristinaKosovo
- National Institute of Public HealthPristinaKosovo
| | | | | | | | | | | |
Collapse
|
44
|
Ruf S, Harding D, Sorie S, Janneh FM, Theuring S. Post-Omicron SARS-CoV-2 serostatus in Sierra Leone: A cross-sectional study in a maternity hospital setting in Freetown, November/December 2022. J Infect Public Health 2024; 17:102518. [PMID: 39142080 DOI: 10.1016/j.jiph.2024.102518] [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: 01/02/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Spread of SARS-CoV-2 in Sub-Saharan African countries has been poorly investigated, especially in the later pandemic stages. We aimed to assess the post-Omicron situation in Sierra Leone in November/December 2022 considering SARS-CoV-2 serostatus, vaccinations, and behavioral factors. METHODS In a cross-sectional study conducted in a maternity hospital in Freetown, Sierra Leone, both patients and staff provided dried blood spot samples for analysis of anti-S and anti-N IgG prevalence using Anti-SARS-CoV-2-ELISA. Additionally, we collected sociodemographic and infection-related information through questionnaires. Outcome parameters included seropositivity, infection-related seroprevalence, and self-reported vaccination status. We used logistic regression to identify associations with prior infection and with vaccination status. RESULTS Out of 791 participants (389 patients, 402 staff), 670 (84.7 %) displayed a positive SARS-CoV-2 serostatus resulting from either infection or vaccination. Among a sub-sample of 514 participants within which determination of prior natural infection was possible, 441individuals (85.8 %) were affected. Prior infection was associated with female sex and tertiary education level. Overall, 60.3 % reported having been vaccinated. Staff as opposed to patients, and individuals with higher socioeconomic status were more likely to report vaccination. Individuals who assessed their risk of COVID-19 as either higher or lower compared to a medium-level risk were more likely to have contracted the virus and less likely to have received vaccination. CONCLUSION Our findings suggest that since the Omicron wave in 2022, the Sierra Leonean population has almost universally been exposed to SARS-CoV-2. While this is encouraging in the light of relatively low excess mortality in the country, future investigations on the long-term effect of high viral exposure on epidemic resilience and public health impact will be crucial.
Collapse
Affiliation(s)
- Sebastian Ruf
- Institute of International Health, Charité- Universitätsmedizin Berlin, Corporate Member of Humboldt University and Freie Universität Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Doris Harding
- Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Samuel Sorie
- Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Foday Musa Janneh
- Ministry of Health and Sanitation, Freetown, Sierra Leone; Princess Christian Maternity Hospital, Freetown, Sierra Leone
| | - Stefanie Theuring
- Institute of International Health, Charité- Universitätsmedizin Berlin, Corporate Member of Humboldt University and Freie Universität Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
45
|
Tancredi S, Cullati S, Chiolero A. Surveillance bias in the assessment of the size of COVID-19 epidemic waves: a case study. Public Health 2024; 234:98-104. [PMID: 38972230 DOI: 10.1016/j.puhe.2024.06.006] [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: 01/29/2024] [Revised: 05/13/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024]
Abstract
OBJECTIVES To estimate the size of COVID-19 waves using four indicators across three pandemic periods and assess potential surveillance bias. STUDY DESIGN Case study using data from one region of Switzerland. METHODS We compared cases, hospitalizations, deaths, and seroprevalence during three periods including the first three pandemic waves (period 1: Feb-Oct 2020; period 2: Oct 2020-Feb 2021; period 3: Feb-Aug 2021). Data were retrieved from the Federal Office of Public Health or estimated from population-based studies. To assess potential surveillance bias, indicators were compared to a reference indicator, i.e. seroprevalence during periods 1 and 2 and hospitalizations during the period 3. Timeliness of indicators (the duration from data generation to the availability of the information to decision-makers) was also evaluated. RESULTS Using seroprevalence (our reference indicator for period 1 and 2), the 2nd wave size was slightly larger (by a ratio of 1.4) than the 1st wave. Compared to seroprevalence, cases largely overestimated the 2nd wave size (2nd vs 1st wave ratio: 6.5), while hospitalizations (ratio: 2.2) and deaths (ratio: 2.9) were more suitable to compare the size of these waves. Using hospitalizations as a reference, the 3rd wave size was slightly smaller (by a ratio of 0.7) than the 2nd wave. Cases or deaths slightly underestimated the 3rd wave size (3rd vs 2nd wave ratio for cases: 0.5; for deaths: 0.4). The seroprevalence was not useful to compare the size of these waves due to high vaccination rates. Across all waves, timeliness for cases and hospitalizations was better than for deaths or seroprevalence. CONCLUSIONS The usefulness of indicators for assessing the size of pandemic waves depends on the type of indicator and the period of the pandemic.
Collapse
Affiliation(s)
- S Tancredi
- Population Health Laboratory (#PopHealthLab), University of Fribourg, Fribourg, Switzerland; Swiss School of Public Health (SSPH+), Zurich, Switzerland.
| | - S Cullati
- Population Health Laboratory (#PopHealthLab), University of Fribourg, Fribourg, Switzerland; Swiss School of Public Health (SSPH+), Zurich, Switzerland; Quality of Care Service, University Hospitals of Geneva, Geneva, Switzerland
| | - A Chiolero
- Population Health Laboratory (#PopHealthLab), University of Fribourg, Fribourg, Switzerland; Swiss School of Public Health (SSPH+), Zurich, Switzerland; School of Population and Global Health, McGill University, Montreal, Canada; Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
| |
Collapse
|
46
|
Meslé MMI, Brown J, Mook P, Katz MA, Hagan J, Pastore R, Benka B, Redlberger-Fritz M, Bossuyt N, Stouten V, Vernemmen C, Constantinou E, Maly M, Kynčl J, Sanca O, Krause TG, Vestergaard LS, Leino T, Poukka E, Gkolfinopoulou K, Mellou K, Tsintziloni M, Molnár Z, Aspelund G, Thordardottir M, Domegan L, Kelly E, O'Donell J, Urdiales AM, Riccardo F, Sacco C, Bumšteinas V, Liausediene R, Mossong J, Vergison A, Borg ML, Melillo T, Kocinski D, Pollozhani E, Meijerink H, Costa D, Gomes JP, Leite PP, Druc A, Gutu V, Mita V, Lazar M, Popescu R, Popovici O, Musilová M, Mrzel M, Socan M, Učakar V, Limia A, Mazagatos C, Olmedo C, Dabrera G, Kall M, Sinnathamby M, McGowan G, McMenamin J, Morrison K, Nitzan D, Widdowson MA, Smallwood C, Pebody R. Estimated number of lives directly saved by COVID-19 vaccination programmes in the WHO European Region from December, 2020, to March, 2023: a retrospective surveillance study. THE LANCET. RESPIRATORY MEDICINE 2024; 12:714-727. [PMID: 39127051 DOI: 10.1016/s2213-2600(24)00179-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/14/2024] [Accepted: 05/24/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND By March, 2023, 54 countries, areas, and territories (hereafter CAT) in the WHO European Region had reported more than 2·2 million COVID-19-related deaths to the WHO Regional Office for Europe. Here, we estimated how many lives were directly saved by vaccinating adults in the WHO European Region from December, 2020, to March, 2023. METHODS In this retrospective surveillance study, we estimated the number of lives directly saved by age group, vaccine dose, and circulating variant-of-concern (VOC) period, regionally and nationally, using weekly data on COVID-19 mortality and infection, COVID-19 vaccination uptake, and SARS-CoV-2 virus characterisations by lineage downloaded from The European Surveillance System on June 11, 2023, as well as vaccine effectiveness data from the literature. We included data for six age groups (25-49 years, 50-59 years, ≥60 years, 60-69 years, 70-79 years, and ≥80 years). To be included in the analysis, CAT needed to have reported both COVID-19 vaccination and mortality data for at least one of the four older age groups. Only CAT that reported weekly data for both COVID-19 vaccination and mortality by age group for 90% of study weeks or more in the full study period were included. We calculated the percentage reduction in the number of expected and reported deaths. FINDINGS Between December, 2020, and March, 2023, in 34 of 54 CAT included in the analysis, COVID-19 vaccines reduced deaths by 59% overall (CAT range 17-82%), representing approximately 1·6 million lives saved (range 1·5-1·7 million) in those aged 25 years or older: 96% of lives saved were aged 60 years or older and 52% were aged 80 years or older; first boosters saved 51% of lives, and 60% were saved during the Omicron period. INTERPRETATION Over nearly 2·5 years, most lives saved by COVID-19 vaccination were in older adults by first booster dose and during the Omicron period, reinforcing the importance of up-to-date vaccination among the most at-risk individuals. Further modelling work should evaluate indirect effects of vaccination and public health and social measures. FUNDING US Centers for Disease Control and Prevention.
Collapse
Affiliation(s)
- Margaux M I Meslé
- World Health Organization Regional Office for Europe, Copenhagen, Denmark.
| | - Jeremy Brown
- World Health Organization Regional Office for Europe, Copenhagen, Denmark
| | - Piers Mook
- World Health Organization Regional Office for Europe, Copenhagen, Denmark
| | - Mark A Katz
- World Health Organization Regional Office for Europe, Copenhagen, Denmark
| | - José Hagan
- World Health Organization Regional Office for Europe, Copenhagen, Denmark
| | - Roberta Pastore
- World Health Organization Regional Office for Europe, Copenhagen, Denmark
| | - Bernhard Benka
- Österreichische Agentur für Gesundheit und Ernährungssicherheit, Vienna, Austria
| | | | | | | | | | | | - Marek Maly
- National Institute of Public Health, Prague, Czechia
| | - Jan Kynčl
- National Institute of Public Health, Prague, Czechia; Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Ondrej Sanca
- Institute of Health Information and Statistics of the Czech Republic, Nové Město, Czechia
| | | | | | - Tuija Leino
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Eero Poukka
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | | | - Kassiani Mellou
- Hellenic National Public Health Organization, Athens, Greece
| | | | - Zsuzsanna Molnár
- National Center for Public Health and Pharmacy, Budapest, Hungary
| | - Gudrun Aspelund
- Centre for Health Security and Communicable Disease Control, Reykjavik, Iceland
| | | | - Lisa Domegan
- Health Service Executive-Health Protection Surveillance Centre, Dublin, Ireland
| | - Eva Kelly
- Health Service Executive-Health Protection Surveillance Centre, Dublin, Ireland
| | - Joan O'Donell
- Health Service Executive-Health Protection Surveillance Centre, Dublin, Ireland
| | | | | | | | - Viktoras Bumšteinas
- National Public Health Center under the Ministry of Health, Vilnius, Lithuania
| | - Rasa Liausediene
- National Public Health Center under the Ministry of Health, Vilnius, Lithuania
| | | | | | - Maria-Louise Borg
- Infectious Disease Prevention and Control Unit, Health Promotion and Disease Prevention Directorate, Pietà, Malta
| | - Tanya Melillo
- Infectious Disease Prevention and Control Unit, Health Promotion and Disease Prevention Directorate, Pietà, Malta
| | - Dragan Kocinski
- Institute of Public Health of Republic of North Macedonia, Skopje, North Macedonia
| | - Enkela Pollozhani
- Institute of Public Health of Republic of North Macedonia, Skopje, North Macedonia
| | | | - Diana Costa
- Directorate of Disease Prevention and Health Promotion, Directorate-General of Health, Lisbon, Portugal
| | - João Paulo Gomes
- National Health Institute Doutor Ricardo Jorge, Lisbon, Portugal
| | - Pedro Pinto Leite
- Directorate of Information and Analysis, Directorate-General of Health, Lisbon, Portugal
| | - Alina Druc
- National Agency for Public Health, Chisinau, Moldova
| | | | - Valentin Mita
- National Agency for Public Health, Chisinau, Moldova
| | - Mihaela Lazar
- Cantacuzino National Military Medical Institute for Research and Development, Bucharest, Romania
| | | | | | | | - Maja Mrzel
- National Institute of Public Health, Ljubljana, Slovenia
| | - Maja Socan
- National Institute of Public Health, Ljubljana, Slovenia
| | | | | | | | | | | | | | | | | | | | | | - Dorit Nitzan
- World Health Organization Regional Office for Europe, Copenhagen, Denmark
| | | | | | - Richard Pebody
- World Health Organization Regional Office for Europe, Copenhagen, Denmark
| |
Collapse
|
47
|
Leon-Rojas JE, Arias-Erazo F, Jiménez-Arias P, Recalde-Navarrete R, Guevara A, Coloma J, Martin M, Chis Ster I, Cooper P, Romero-Sandoval N, on behalf of the Seroprevalence ECU-Group. COVID-19 IgG seropositivity and its determinants in occupational groups of varying infection risks in two Andean cities of Ecuador before mass vaccination. PLoS One 2024; 19:e0309466. [PMID: 39208200 PMCID: PMC11361580 DOI: 10.1371/journal.pone.0309466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic has caused over 68.7 million infections and 1.35 million deaths in South America. There are limited data on SARS-CoV-2 seropositivity and its determinants from Andean countries prior to mass vaccinations against COVID-19. OBJECTIVE To estimate SARS-CoV-2 seropositivity and its determinants before vaccination in occupational groups of adults presumed to have different levels of exposure and associations with potential symptomatology. METHODS We measured seropositivity of anti-SARS-CoV-2 IgG antibodies in a cross-sectional study of vaccine-naïve adults aged 18 years and older, recruited within three occupational risk groups (defined as low [LR], moderate [MR], and high [HR]) between January and September 2021 in two Andean cities in Ecuador. Associations with risk factors were estimated using logistic regression. RESULTS In a sample of 882 adults, IgG seropositivity for the three different occupational risk groups was 39.9% (CI 95% 35.3-44.6), 74.6% (CI 95% 66.4-81.4), and 39.0% (CI 95% 34.0-44.4) for the HR, MR, and LR groups, respectively. History of an illness with loss of taste and/or smell was significantly associated with seropositivity in all occupational groups, with adjusted ORs of 14.31 (95%CI, 5.83-35.12; p<0.001), 14.34 (95%CI 3.01-68.42; p<0.001), and 8.79 (95%CI 2.69-28.72; p<0.001), for the HR, MR, and LR groups, respectively; while fever was significant for the LR group with an adjusted OR of 1.24 (95%CI, 1.11-4.57; p = 0.025) and myalgia for the HR group with an adjusted OR of 2.07 (95%CI, 1.13-3.81; p = 0.019). CONCLUSION Notable proportions of seropositivity were seen in all occupational groups between January and September 2021 prior to mass vaccination. Loss of taste and/or smell was strongly associated with presence of anti-SARS-CoV-2 IgG antibodies irrespective of presumed occupational exposure risk.
Collapse
Affiliation(s)
- Jose E. Leon-Rojas
- Departamento de Pediatría, Obstetricia y Ginecología y Medicina Preventiva, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain
- Research Network Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL), Universidad Internacional del Ecuador, Quito, Ecuador
| | - Fernanda Arias-Erazo
- School of Medicine, Universidad Internacional del Ecuador, Quito, Ecuador
- Grupo de Investigación en Sanidad Animal y Humana (GISAH) ESPE, Quito, Ecuador
| | - Patricia Jiménez-Arias
- Research Network Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL), Universidad Internacional del Ecuador, Quito, Ecuador
- Grupo de Investigación en Sanidad Animal y Humana (GISAH) ESPE, Quito, Ecuador
- Departamento de Ciencias de la Vida y de la Agricultura, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Ecuador
| | - Ricardo Recalde-Navarrete
- Research Network Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL), Universidad Internacional del Ecuador, Quito, Ecuador
- Medical School, Universidad Tecnica de Ambato, Ambato, Ecuador
| | | | - Josefina Coloma
- School of Public Health, University of California, Berkeley, Berkeley, CA, United States of America
| | - Miguel Martin
- Departamento de Pediatría, Obstetricia y Ginecología y Medicina Preventiva, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain
- Research Network Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL), Universidad Internacional del Ecuador, Quito, Ecuador
- School of Medicine, Universidad Internacional del Ecuador, Quito, Ecuador
| | - Irina Chis Ster
- Institute of Infection and Immunity, St George’s University of London, London, United Kingdom
| | - Philip Cooper
- School of Medicine, Universidad Internacional del Ecuador, Quito, Ecuador
- Institute of Infection and Immunity, St George’s University of London, London, United Kingdom
| | - Natalia Romero-Sandoval
- Research Network Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL), Universidad Internacional del Ecuador, Quito, Ecuador
- School of Medicine, Universidad Internacional del Ecuador, Quito, Ecuador
| | | |
Collapse
|
48
|
Tess BH, Turchi Martelli CM, Goi Porto Alves MC, Cortes F, Ivata Bernal RT, Vieira de Souza W, de Albuquerque Luna EJ, da Cunha Rodrigues L, Cavallari Nunes M, de Castro Reinach F, Granato CFH, Rizzatti EG, Tostes Pintão MC. Seroprevalence trends of anti-SARS-CoV-2 antibodies in the adult population of the São Paulo Municipality, Brazil: Results from seven serosurveys from June 2020 to April 2022. The SoroEpi MSP Study. PLoS One 2024; 19:e0309441. [PMID: 39186722 PMCID: PMC11346932 DOI: 10.1371/journal.pone.0309441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/07/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND Sequential population-based household serosurveys of SARS-CoV-2 covering the COVID-19 pre- and post-vaccination periods are scarce in Brazil. This study investigated seropositivity trends in the municipality of São Paulo. METHODS We conducted seven cross-sectional surveys of adult population-representative samples between June 2020 and April 2022. The study design included probabilistic sampling, test for SARS-CoV-2 antibodies using the Roche Elecsys anti-nucleocapsid assay, and statistical adjustments for population demographics and non-response. The weighted seroprevalences with 95% confidence intervals (CI) were estimated by sex, age group, race, schooling, and mean income study strata. Time trends in seropositivity were assessed using the Joinpoint model. We compared infection-induced seroprevalences with COVID-19 reported cases in the pre-vaccination period. RESULTS The study sample comprised 8,134 adults. The overall SARS-CoV-2 seroprevalence increased from 11.4% (95%CI: 9.2-13.6) in June 2020 to 24.9% (95%CI: 21.0-28.7) in January 2021; from 38.1% (95%CI: 34.3-41.9) in April 2021 to 77.7% (95%CI: 74.4-81.0) in April 2022. The prevalence over time was higher in the subgroup 18-39 years old than in the older groups from Survey 3 onwards. The self-declared Black or mixed (Pardo) group showed a higher prevalence in all surveys compared to the White group. Monthly prevalence rose steeply from January 2021 onwards, particularly among those aged 60 years or older. The infection-to-case ratios ranged from 8.9 in June 2020 to 4.3 in January 2021. CONCLUSIONS The overall seroprevalence rose significantly over time and with age and race subgroup variations. Increases in the 60 years or older age and the White groups were faster than in younger ages and Black or mixed (Pardo) race groups in the post-vaccination period. Our data may add to the understanding of the complex and changing population dynamics of the SARS-CoV-2 infection, including the impact of vaccination strategies and the modelling of future epidemiological scenarios.
Collapse
Affiliation(s)
- Beatriz Helena Tess
- Departamento de Medicina Preventiva, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | | | | | - Fanny Cortes
- Programa de Pós-Graduação em Ciências da Saúde, Universidade de Pernambuco, Recife, PE, Brasil
| | - Regina Tomie Ivata Bernal
- Programa de Pós-Graduação, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | | | | | - Laura da Cunha Rodrigues
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | | | | | - Edgar Gil Rizzatti
- Divisão de Pesquisa e Desenvolvimento, Grupo Fleury, São Paulo, SP, Brasil
| | | |
Collapse
|
49
|
Ramírez-Santana M, Correa J, Núñez Franz L, Apablaza M, Rubilar P, Vial C, Jimena Cortes L, Hormazábal J, Canales L, Vial P, Aguilera X. Overcoming Health Inequities: Spatial Analysis of Seroprevalence and Vaccination Against COVID-19 in Chile. Health Equity 2024; 8:558-567. [PMID: 40125367 PMCID: PMC11392677 DOI: 10.1089/heq.2023.0204] [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] [Accepted: 02/06/2024] [Indexed: 03/25/2025] Open
Abstract
Background In unequal economies, the spread of the first waves of the COVID-19 was usually associated with low socioeconomic status of individuals and their families. Chile exemplified this. By mid-2020, Chile had one of the highest SARS-CoV-2 infection rates in the world predominantly in poorer areas. A year later, the country launched a universal vaccination campaign based on the national strategy of immunization established in 1975. By 2022, Chile presented one of the highest COVID-19 vaccination coverages globally, reaching 94.3% of the population with the primary scheme by the end of 2022. Objective This study analyzes the spatial distribution of SARS-CoV-2 seroprevalence at the beginning of the pandemic (2020) compared with the seroprevalence after 2 years of ongoing epidemic and COVID-19 vaccination campaigns (2022). Methods Two population-based random samples of individuals aged 7 years and older from two Chilean cities were studied. Utilizing an enzyme-linked immunosorbent assay test, IgG antibodies were measured in serum of 1061 participants in 2020, and 853 in 2022. Results Using the Global Moran's Index, the seroprevalence distribution pattern for the year 2020 showed clustering in the two cities. Conversely, seroprevalence and vaccinations were homogeneously distributed in 2022. These results show the success of the vaccination campaign in Chile, not only in coverage but also because it widely reached all individuals. Conclusions The uptake of this preventive measure is high, regardless of the social and economic factors, achieving broad population immunity. The extensive deployment of the primary health care network contributed to reducing health inequities and promoting to universal health access.
Collapse
Affiliation(s)
- Muriel Ramírez-Santana
- Departamento de Salud Pública, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Juan Correa
- Centro Producción del Espacio, Universidad de Las Américas, Santiago, Chile
- Doctorado en Geografía, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Loreto Núñez Franz
- Departamento de Salud Pública, Facultad de Ciencias de la Salud, Universidad de Talca, Talca, Chile
| | | | - Paola Rubilar
- Centro de Epidemiología y Políticas de Salud, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Cecilia Vial
- Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Lina Jimena Cortes
- Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Juan Hormazábal
- Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Luis Canales
- Facultad de Economía y Negocios, Universidad de Talca, Talca, Chile
| | - Pablo Vial
- Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Ximena Aguilera
- Centro de Epidemiología y Políticas de Salud, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| |
Collapse
|
50
|
Tunheim G, Fossum E, Robertson AH, Rø GØI, Chopra A, Vaage JT, Vikse EL, Kran AMB, Magnus P, Trogstad L, Mjaaland S, Hungnes O, Lund-Johansen F. Characterization of the SARS-CoV-2 antibody landscape in Norway in the late summer of 2022: high seroprevalence in all age groups with patterns of primary Omicron infection in children and hybrid immunity in adults. BMC Infect Dis 2024; 24:841. [PMID: 39164637 PMCID: PMC11334563 DOI: 10.1186/s12879-024-09670-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND According to Norwegian registries, 91% of individuals ≥ 16 years had received ≥ 1 dose of COVID-19 vaccine by mid-July 2022, whereas less than 2% of children < 12 years were vaccinated. Confirmed COVID-19 was reported for 27% of the population, but relaxation of testing lead to substantial underreporting. We have characterized the humoral immunity to SARS-CoV-2 in Norway in the late summer of 2022 by estimating the seroprevalence and identifying antibody profiles based on reactivity to Wuhan or Omicron-like viruses in a nationwide cross-sectional collection of residual sera, and validated our findings using cohort sera. METHODS 1,914 anonymized convenience sera and 243 NorFlu-cohort sera previously collected from the Oslo-area with reported infection and vaccination status were analyzed for antibodies against spike, the receptor-binding domain (RBD) of the ancestral Wuhan strain and Omicron BA.2 RBD, and nucleocapsid (N). Samples were also tested for antibodies inhibiting RBD-ACE2 interaction. Neutralization assays were performed on subsets of residual sera against B.1, BA.2, XBB.1.5 and BQ.1.1. RESULTS The national seroprevalence estimate from vaccination and/or infection was 99.1% (95% CrI 97.0-100.0%) based on Wuhan (spike_W and RBD_W) and RBD_BA2 antibodies. Sera from children < 12 years had 2.2 times higher levels of antibodies against RBD_BA2 than RBD_W and their seroprevalence estimate showed a 14.4 percentage points increase when also including anti-RBD_BA2 antibodies compared to Wuhan-antibodies alone. 50.3% (95% CI 45.0-55.5%) of residual sera from children and 38.1% (95% CI 36.0-40.4%) of all residual sera were positive for anti-N-antibodies. By combining measurements of binding- and ACE2-RBD-interaction-inhibiting antibodies, reactivity profiles indicative of infection and vaccination history were identified and validated using cohort sera. Residual sera with a profile indicative of hybrid immunity were able to neutralize newer Omicron variants XBB.1.5 and BQ.1.1. CONCLUSIONS By late summer of 2022, most of the Norwegian population had antibodies to SARS-CoV-2, and almost all children had been infected. Antibody profiles indicated that children mostly had experienced a primary Omicron infection, while hybrid immunity was common among adults. The finding that sera displaying hybrid immunity could neutralize newer Omicron variants indicates that Wuhan-like priming of the immune response did not have a harmful imprinting effect and that infections induce cross-reacting antibodies against future variants.
Collapse
Affiliation(s)
- Gro Tunheim
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway.
| | - Even Fossum
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Anna Hayman Robertson
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | | | - Adity Chopra
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - John T Vaage
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Elisabeth Lea Vikse
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Anne-Marte Bakken Kran
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Per Magnus
- Center for Fertility and Health, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Lill Trogstad
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Siri Mjaaland
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Olav Hungnes
- Division of Infection Control, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | | |
Collapse
|