1
|
Daher IP, Almeida BDS, de Souza-Silva GA, Marques RF, Soares GHC, Andreata-Santos R, Moretti A, de Oliveira Silva M, Schuch V, Sasahara GL, Kuramoto A, Yamamoto M, Ferreira LCDS, Santos K, Coelho VPCV, Kalil J, Rosa DS, Cunha-Neto E, Boscardin SB. Neutralizing antibody responses after a two-dose regimen with BNT162b2, CoronaVac or ChAdOx1-S in Brazil: Differential neutralization of SARS-CoV-2 omicron variants. Clin Immunol 2025; 276:110492. [PMID: 40185297 DOI: 10.1016/j.clim.2025.110492] [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: 01/03/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
The emergence of SARS-CoV-2 variants has reduced antibody effectiveness, affecting vaccine protection. This study evaluated neutralizing antibodies against Wuhan strain and several variants, including Alpha, Beta, Gamma, Delta, and Omicron, in Brazilians vaccinated twice with CoronaVac, ChAdOx1-S, or BNT162b2 before Delta and Omicron emerged. After the booster, strong antibody responses to the Wuhan strain were seen in all groups, but BNT162b2 resulted in higher anti-Spike and anti-RBD IgG levels. While all vaccines showed some cross-neutralization against Alpha, Beta, Gamma, and Delta, only BNT162b2 was effective against Omicron BA.2 and BA.4/5 subvariants. Furthermore, BNT162b2 vaccination showed a positive correlation between Wuhan RBD-specific IgG and Omicron neutralizing antibodies. This group demonstrated distinct clustering patterns of neutralizing antibodies against all variants, unlike those from CoronaVac and ChAdOx1-S. The findings suggest BNT162b2 offers broader neutralization capability, highlighting the benefit of booster shots with bivalent mRNA vaccines to enhance immune responses against emerging variants.
Collapse
Affiliation(s)
- Isabela Pazotti Daher
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil; Instituto Ciências Biomédicas da Universidade de São Paulo, ICB-USP, São Paulo 05508-000, Brazil
| | - Bianca da Silva Almeida
- Instituto Ciências Biomédicas da Universidade de São Paulo, ICB-USP, São Paulo 05508-000, Brazil
| | | | - Rodolfo Ferreira Marques
- Instituto Ciências Biomédicas da Universidade de São Paulo, ICB-USP, São Paulo 05508-000, Brazil
| | | | - Robert Andreata-Santos
- Instituto Ciências Biomédicas da Universidade de São Paulo, ICB-USP, São Paulo 05508-000, Brazil
| | - Ana Moretti
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | | | - Viviane Schuch
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Greyce Luri Sasahara
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Andréia Kuramoto
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Marcio Yamamoto
- Instituto Ciências Biomédicas da Universidade de São Paulo, ICB-USP, São Paulo 05508-000, Brazil
| | | | - Keity Santos
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil; Instituto de Investigação em Imunologia-Instituto Nacional de Ciências e Tecnologia (iii-INCT), São Paulo 05403-000, Brazil
| | - Verônica P C V Coelho
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil; Instituto de Investigação em Imunologia-Instituto Nacional de Ciências e Tecnologia (iii-INCT), São Paulo 05403-000, Brazil
| | - Jorge Kalil
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil; Instituto de Investigação em Imunologia-Instituto Nacional de Ciências e Tecnologia (iii-INCT), São Paulo 05403-000, Brazil
| | - Daniela Santoro Rosa
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil; Instituto de Investigação em Imunologia-Instituto Nacional de Ciências e Tecnologia (iii-INCT), São Paulo 05403-000, Brazil
| | - Edecio Cunha-Neto
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil; Instituto de Investigação em Imunologia-Instituto Nacional de Ciências e Tecnologia (iii-INCT), São Paulo 05403-000, Brazil.
| | - Silvia Beatriz Boscardin
- Instituto Ciências Biomédicas da Universidade de São Paulo, ICB-USP, São Paulo 05508-000, Brazil; Instituto de Investigação em Imunologia-Instituto Nacional de Ciências e Tecnologia (iii-INCT), São Paulo 05403-000, Brazil.
| |
Collapse
|
2
|
Chakraborty C, Lo YH, Bhattacharya M, Das A, Wen ZH. Looking beyond the origin of SARS-CoV-2: Significant strategic aspects during the five-year journey of COVID-19 vaccine development. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102527. [PMID: 40291378 PMCID: PMC12032352 DOI: 10.1016/j.omtn.2025.102527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
It has been five years since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and we are also approaching the five-year mark of the COVID-19 pandemic. The vaccine is a significant weapon in combating infectious diseases like SARS-CoV-2. Several vaccines were developed against SARS-CoV-2, and they demonstrated efficacy and safety during these five years. The rapid development of multiple next-generation vaccine candidates in different platforms with very little time is the success story of the vaccine development endeavor. This remarkable success of rapid vaccine development is a new paradigm for fast vaccine development that might help develop infectious diseases and fight against the pandemic. With the completion of five years since the beginning of SARS-CoV-2 origin, we are looking back on the five years and reviewing the milestones, vaccine platforms, animal models, clinical trials, successful collaborations, vaccine safety, real-world effectiveness, and challenges. Lessons learned during these five years will help us respond to public health emergencies and to fight the battle against future pandemics.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Yi-Hao Lo
- Department of Family Medicine, Zuoying Armed Forces General Hospital, Kaohsiung 81342, Taiwan
- Department of Nursing, Meiho University, Neipu Township, Pingtung County 91200, Taiwan
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, Odisha 756020, India
| | - Arpita Das
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, #70 Lien-Hai Road, Kaohsiung 804201, Taiwan
- National Museum of Marine Biology & Aquarium, # 2 Houwan Road, Checheng, Pingtung 94450, Taiwan
| |
Collapse
|
3
|
Morgenstern C, Rawson T, Hinsley W, Perez Guzman PN, Bhatt S, Ferguson NM. Socioeconomic and temporal heterogeneity in SARS-CoV-2 exposure and disease in England from May 2020 to February 2023. SCIENCE ADVANCES 2025; 11:eadu8678. [PMID: 40397734 PMCID: PMC12094233 DOI: 10.1126/sciadv.adu8678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/16/2025] [Indexed: 05/23/2025]
Abstract
The impact of COVID-19 varied significantly by deprivation, ethnicity, and policy measures. We analyzed individual-level data on 12,310,485 first SARS-CoV-2 Pillar 2-PCR-confirmed infections, 439,083 hospitalizations, 107,823 deaths, and vaccination records in England from May 2020 to February 2022. Poisson regression models adjusted for demographic and temporal factors showed higher incidence rate ratios (IRRs) for severe outcomes in the most deprived areas compared to the least. We note higher IRRs for severe outcomes for all non-white relative to white ethnicities. The magnitude of IRRs for both deprivation and ethnicities declined from the wild-type to the omicron periods for severe outcomes. For infections, we observed IRRs above one for non-white ethnicities during the wild-type and alpha periods. Vaccination significantly reduced risks across all groups. For severe outcomes, preexisting health inequalities led to large and persistent disparities. For infections, measures must be structured with ethnicity and deprivation in mind early in a pandemic.
Collapse
Affiliation(s)
- Christian Morgenstern
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Thomas Rawson
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Wes Hinsley
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Pablo N. Perez Guzman
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Samir Bhatt
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
- University of Copenhagen, Copenhagen, Denmark
| | - Neil M. Ferguson
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| |
Collapse
|
4
|
Kumar N, Segovia D, Kumar P, Atti HB, Kumar S, Mishra J. Mucosal implications of oral Jak3-targeted drugs in COVID patients. Mol Med 2025; 31:203. [PMID: 40410684 PMCID: PMC12100796 DOI: 10.1186/s10020-025-01260-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025] Open
Abstract
The JAK family, particularly JAK3, plays a crucial role in immune signaling and inflammatory responses. Dysregulated JAK3 activation in SARS-CoV-2 infections has been associated with severe inflammation and respiratory complications, making JAK inhibitors a viable therapeutic option. However, their use raises concerns regarding immunosuppression, which could increase susceptibility to secondary infections. While long-term adverse effects are less of a concern in acute COVID-19 treatment, patient selection and monitoring remain critical. Furthermore, adverse effects associated with oral JAK3 inhibitors necessitate the exploration of alternative strategies to optimize therapeutic efficacy while minimizing risks. This review highlights the role of JAK3 in immune and epithelial cells, examines the adverse effects of oral JAK3 inhibitors in COVID-19 and other treatments, and discusses alternative therapeutic strategies for improving patient outcomes.
Collapse
Affiliation(s)
- Narendra Kumar
- ILR-College of Pharmacy, Texas A&M University Health Science Center, Kingsville, TX, USA.
| | - Daniel Segovia
- ILR-College of Pharmacy, Texas A&M University Health Science Center, Kingsville, TX, USA
| | - Priyam Kumar
- University of Pennsylvania, Philadelphia, PA, USA
| | - Hima Bindu Atti
- ILR-College of Pharmacy, Texas A&M University Health Science Center, Kingsville, TX, USA
| | - Soaham Kumar
- Veterans Memorial High School, Corpus Christi, TX, USA
| | - Jayshree Mishra
- ILR-College of Pharmacy, Texas A&M University Health Science Center, Kingsville, TX, USA.
| |
Collapse
|
5
|
Hasan Z, Masood KI, Qaiser S, Kanji A, Mwenda F, Alenquer M, Iqbal J, Ferreira F, Wassan Y, Balouch S, Yameen M, Hussain S, Begum K, Feroz K, Muhammad S, Sadiqa A, Akhtar M, Habib A, Ahmed SMA, Mian AA, Hussain R, Amorim MJ, Bhutta ZA. Comparative study of humoral and cellular immunity against SARS-CoV-2 induced by different COVID-19 vaccine types: Insights into protection against wildtype, Delta and JN.1 omicron strains. Vaccine 2025; 59:127270. [PMID: 40408899 DOI: 10.1016/j.vaccine.2025.127270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/19/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025]
Abstract
We investigated the effectiveness of different COVID-19 vaccinations administered in Pakistan by studying the effect of inactivated virus, mRNA and vector formulations. This study in 916 participants was conducted between October 2021 and July 2022. Subjects receiving inactivated (A), mRNA (B), one-dose vector (C), and two-dose vector (D) vaccines were sampled at baseline, 6, 12, and 24 weeks. Serum IgG antibodies to wildtype Spike and its receptor binding domain (RBD) were measured. Pseudovirus particle-based neutralizing assays against wildtype, Delta, and JN.1 variants were performed. T cell IFN-γ responses to SARS-CoV-2 antigens were measured. Participants were aged 37.05 ± 14.44 years and comprised 48.6 % females. Baseline Spike seropositivity rose from 90 % to 96 % by 24 weeks; and 40 % to 90 % against RBD. Group B participants had the highest anti-RBD levels which peaked by 6 weeks. IgG RBD in group A and C increased up until 24 weeks. Anti-RBD levels were reduced in those over 50 years. At baseline neutralizing titers were present at 38.5 % against wildtype and in 34.2 % against Delta variants. Titers doubled in vaccine groups A-C by 12 weeks, with highest titers in B and lowest in group C participants. At baseline, neutralizing titers against the JN.1 variant were absent but low titers were evident in 10 % of participants after 12 weeks. T cell reactivity to SARS-CoV-2 increased from 31 % at baseline to 50 % in group A and 73 % in group B participants by 6 weeks after vaccination. Presence of immunity against wildtype and Delta variants in one-third of participants at baseline could be due to sub-clinical infections. Increase in humoral and cellular immunity was greater after mRNA as compared with inactivated vaccinations. As COVID-19 morbidity in the population remained low, our data supports effectiveness of multiple vaccine formulations in protecting against severe COVID-19 in this high transmission population.
Collapse
Affiliation(s)
- Zahra Hasan
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan.
| | - Kiran Iqbal Masood
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Shama Qaiser
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Akbar Kanji
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Fridah Mwenda
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Marta Alenquer
- Católica Biomedical Research Centre, Católica Medical School, Universidade Católica Portuguesa, Palma de Cima, 1649-023 Lisboa, Portugal
| | - Junaid Iqbal
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | - Filipe Ferreira
- Católica Biomedical Research Centre, Católica Medical School, Universidade Católica Portuguesa, Palma de Cima, 1649-023 Lisboa, Portugal
| | - Yaqub Wassan
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | - Sadaf Balouch
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Maliha Yameen
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Shahneel Hussain
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | - Kehkashan Begum
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | - Khalid Feroz
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | - Sajid Muhammad
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | - Ayesha Sadiqa
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Mishgan Akhtar
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Atif Habib
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan
| | | | - Afsar Ali Mian
- Center for Regenerative Medicine, AKU, Karachi, Pakistan
| | - Rabia Hussain
- Department of Pathology and Laboratory Medicine(1), The Aga Khan University (AKU), Karachi, Pakistan
| | - Maria Joao Amorim
- Católica Biomedical Research Centre, Católica Medical School, Universidade Católica Portuguesa, Palma de Cima, 1649-023 Lisboa, Portugal
| | - Zulfiqar A Bhutta
- Center of Excellence in Women and Child Health, AKU, Karachi, Pakistan; Centre for Global Child Health, Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
6
|
Araya S, Lovsin Barle E, Wiesner L, Blum K, Hashimoto K, Fisher C, Schwind M, Galati G, Sehner C, Pfister T, Witzigmann D. RNA therapeutics-An evaluation of potential occupational health hazards and a strategy to establish occupational exposure limits (OELs). JOURNAL OF OCCUPATIONAL AND ENVIRONMENTAL HYGIENE 2025:1-20. [PMID: 40372247 DOI: 10.1080/15459624.2025.2485080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
RNA therapeutics represent a rapidly expanding and innovative group of pharmaceuticals. These new modalities necessitate the establishment of Occupational Exposure Limits (OELs) to ensure safe occupational handling. While there is an established methodology for setting OELs for small molecule therapeutics, this methodology is not readily applicable to large molecule RNA therapeutics that deserve additional considerations in their safety assessment, particularly for aspects related to their unique modes of action. This research, which involves an extensive review of the data available for RNA therapeutics to derive substance-specific OELs and to propose a strategy for low-characterized RNA therapeutics, fills this crucial gap. It is recommended to apply an activity correction factor (ACF) in the OEL formula for large molecules, as representative of the "α" in the OEL formula for small molecules, considering differences in route of administration, critical effects, mechanism of action, and the RNA delivery platform. Additionally, it is proposed to consider lower OEL values for mRNA vaccines as compared to other RNA therapeutics. Finally, it is suggested that the exposure assessment experience that has already been acquired when handling therapeutic proteins can also be used to define containment strategies for RNA therapeutics.
Collapse
Affiliation(s)
- S Araya
- Lonza Group Ltd, Basel, Switzerland
| | - E Lovsin Barle
- Takeda Pharmaceuticals International AG, Glattpark-Opfikon, Switzerland
| | | | - K Blum
- GlaxoSmithKline GmbH & Co. KG, Munich, Germany
| | - K Hashimoto
- Takeda Pharmaceutical Company, Ltd, Fujisawa, Japan
| | - C Fisher
- Takeda Pharmaceuticals International AG, Glattpark-Opfikon, Switzerland
| | - M Schwind
- Sanofi-Aventis Deutschland GmbH, Frankfurt, Germany
| | - G Galati
- Thermo Fisher Scientific, Mississauga, Canada
| | - C Sehner
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - T Pfister
- Hoffmann-La Roche AG, Basel, Switzerland
| | | |
Collapse
|
7
|
Gonzalez Dorta H, Verbeeck J, Crevecoeur J, Morales DR, Loedy N, Cohet C, Willem L, Molenberghs G, Hens N, Kurz X, Quinten C, Abrams S. Utilising the Benefit Risk Assessment of Vaccines (BRAVE) toolkit to evaluate the benefits and risks of Vaxzevria in the EU: a population-based study. Lancet Digit Health 2025; 7:100861. [PMID: 40204579 DOI: 10.1016/j.landig.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/03/2024] [Accepted: 02/05/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Several COVID-19 vaccines have been licensed. To support the assessment of safety signals, we developed a toolkit to support COVID-19 vaccine monitoring and benefit-risk assessment. We aim to show the application of our toolkit in the EU using thrombosis with thrombocytopenia syndrome (TTS) associated with the Vaxzevria (AstraZeneca) vaccine as a use case. METHODS In this population-based study, we used a model incorporating data from multiple EU sources such as The European Surveillance System and EudraVigilance, and estimated the benefits of COVID-19 vaccines by comparing the observed COVID-19 confirmed cases, hospitalisations, intensive care unit (ICU) admissions, and deaths across Europe to the expected numbers in the absence of Vaxzevria vaccination. Risks of TTS associated with Vaxzevria were calculated by comparing the observed number of TTS events in individuals who received Vaxzevria to the expected number of events based on background incidence rates. To visualise the results, we developed a toolkit with an interactive web application. FINDINGS 62 598 505 Vaxzevria vaccines (32 763 183 to females and 29 835 322 to males) had been administered in Europe by Feb 10, 2021. Our results showed that a first dose of Vaxzevria provided benefits across all age groups. Based on vaccine effectiveness estimates and reported coverage in Europe, from Dec 13, 2020 to Dec 31, 2021, vaccination with Vaxzevria was estimated to prevent (per 100 000 doses) 12 113 COVID-19 cases, 1140 hospitalisations, 184 ICU admissions, and 261 deaths. Women aged 30-59 years and males aged 20-29 years had the highest frequency of TTS events. The benefits of vaccination outweighed the risks of TTS in all age groups, with the highest benefits and risks observed in individuals aged 60-69 years. INTERPRETATION Our toolkit and underlying model contextualised the risk of TTS associated with Vaxzevria relative to its benefits. The methodology employed could be applied to other serious adverse events related to COVID-19 or other vaccines. The adaptability and versatility of such toolkits might contribute to strengthening preparedness for future public health emergencies. FUNDING European Medicines Agency.
Collapse
Affiliation(s)
| | - Johan Verbeeck
- Data Science Institute, I-BioStat, Universiteit Hasselt, Hasselt, Belgium
| | - Jonas Crevecoeur
- Data Science Institute, I-BioStat, Universiteit Hasselt, Hasselt, Belgium; I-BioStat, KU Leuven, Leuven, Belgium
| | - Daniel R Morales
- European Medicines Agency, Amsterdam, Netherlands; Division of Population Health and Genomics, University of Dundee, Dundee, UK
| | - Neilshan Loedy
- Data Science Institute, I-BioStat, Universiteit Hasselt, Hasselt, Belgium
| | | | - Lander Willem
- Centre for Health Economics Research and Modelling of Infectious Diseases, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Geert Molenberghs
- Data Science Institute, I-BioStat, Universiteit Hasselt, Hasselt, Belgium; I-BioStat, KU Leuven, Leuven, Belgium
| | - Niel Hens
- Data Science Institute, I-BioStat, Universiteit Hasselt, Hasselt, Belgium; Centre for Health Economics Research and Modelling of Infectious Diseases, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Xavier Kurz
- European Medicines Agency, Amsterdam, Netherlands
| | | | - Steven Abrams
- Data Science Institute, I-BioStat, Universiteit Hasselt, Hasselt, Belgium; Global Health Institute, Family Medicine and Population Health, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
8
|
Connolly A, Noone P, Buggy C, Costello E, Wright F, Farrell C, Lenehan G, Gillen M, Coulahan P, Wall P, McDonnell P, Flavin N. Strategies for Managing the COVID-19 Pandemic and Lessons Learnt: An Irish Perspective. Public Health Rev 2025; 46:1607427. [PMID: 40365087 PMCID: PMC12069057 DOI: 10.3389/phrs.2025.1607427] [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: 04/26/2024] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Background The World Health Organization (WHO) announced the end of the emergency phase of Corona Virus Disease 2019 (COVID-19) in May 2023. Nations across the world address the effects of the pandemic and the need to plan for future pandemics. Ireland heavily focused on isolation and social distancing to curb the infection rate early in the pandemic. These long and extended lockdowns gave Ireland a very unique experience during the COVID-19 pandemic. Analysis Ireland's COVID-19 pandemic management was discussed by an expert panel on the strengths in our national pandemic action plans, areas not sufficiently addressed and requirements to ensure future pandemic preparedness plans are robust. Policy Options Recommendations include having a more strategic plan to protect society's most vulnerable people, a flexible national policy that swiftly implements advances in scientific knowledge and good practices, a robust communication plan including localised information to prevent "pandemic fatigue," and address challenges from restrictions, lockdowns and isolation such as mental health and wellbeing. Conclusion Lessons learnt from the Irish COVID-19 pandemic experience can be utilised for pandemic preparedness plans, nationally and internationally.
Collapse
Affiliation(s)
- Alison Connolly
- UCD Centre for Safety and Health at Work, School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Dublin, Ireland
| | - Peter Noone
- Regional Occupational Health Service, HSE Dublin North East, Naas, Ireland
| | - Conor Buggy
- UCD Centre for Safety and Health at Work, School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Dublin, Ireland
| | - Edel Costello
- Department of Environmental Science, Faculty of Science, Atlantic Technological University Sligo, Sligo, Ireland
| | | | | | | | - Michael Gillen
- BioPharmaChem Ireland, Ibec Head Office, Dublin, Ireland
| | - Peter Coulahan
- UCD Risk Management Office, University College Dublin, Dublin, Ireland
| | - Patrick Wall
- UCD-Centre for Food Safety, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland
| | | | - Nuala Flavin
- Occupational Hygiene Society of Ireland, Meath, Ireland
| |
Collapse
|
9
|
Saha A, Ghosh Roy S, Dwivedi R, Tripathi P, Kumar K, Nambiar SM, Pathak R. Beyond the Pandemic Era: Recent Advances and Efficacy of SARS-CoV-2 Vaccines Against Emerging Variants of Concern. Vaccines (Basel) 2025; 13:424. [PMID: 40333293 PMCID: PMC12031379 DOI: 10.3390/vaccines13040424] [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: 02/17/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
Vaccination has been instrumental in curbing the transmission of SARS-CoV-2 and mitigating the severity of clinical manifestations associated with COVID-19. Numerous COVID-19 vaccines have been developed to this effect, including BioNTech-Pfizer and Moderna's mRNA vaccines, as well as adenovirus vector-based vaccines such as Oxford-AstraZeneca. However, the emergence of new variants and subvariants of SARS-CoV-2, characterized by enhanced transmissibility and immune evasion, poses significant challenges to the efficacy of current vaccination strategies. In this review, we aim to comprehensively outline the landscape of emerging SARS-CoV-2 variants of concern (VOCs) and sub-lineages that have recently surfaced in the post-pandemic years. We assess the effectiveness of existing vaccines, including their booster doses, against these emerging variants and subvariants, such as BA.2-derived sub-lineages, XBB sub-lineages, and BA.2.86 (Pirola). Furthermore, we discuss the latest advancements in vaccine technology, including multivalent and pan-coronavirus approaches, along with the development of several next-generation coronavirus vaccines, such as exosome-based, virus-like particle (VLP), mucosal, and nanomaterial-based vaccines. Finally, we highlight the key challenges and critical areas for future research to address the evolving threat of SARS-CoV-2 subvariants and to develop strategies for combating the emergence of new viral threats, thereby improving preparedness for future pandemics.
Collapse
Affiliation(s)
- Ankita Saha
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
| | - Sounak Ghosh Roy
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Naval Medical Research Command, Silver Spring, MD 20910, USA;
| | - Richa Dwivedi
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN 37208, USA;
| | - Prajna Tripathi
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Kamal Kumar
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA;
| | - Shashank Manohar Nambiar
- Division of Hepatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
10
|
Morris JS. Tracking vaccine effectiveness in an evolving pandemic, countering misleading hot takes and epidemiologic fallacies. Am J Epidemiol 2025; 194:898-907. [PMID: 39218423 PMCID: PMC11978612 DOI: 10.1093/aje/kwae280] [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: 06/09/2023] [Revised: 07/24/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
With the emergence of Omicron during the pandemic and the establishment of antibody waning over time, vaccine effectiveness, especially against infection, declined sharply from the original levels seen after the initial rollout. However, studies have demonstrated that they still provided substantial protection vs severe/fatal disease even with Omicron and after waning. Social media has been rife with reports claiming vaccines provided no benefit and some even claiming they made things worse, often driven by simple presentations of raw observational data using erroneous arguments involving epidemiologic fallacies including the base rate fallacy, Simpson's paradox, and the ecological fallacy and ignoring the extensive bias especially from confounding that is an inherent feature of these data. Similar fallacious arguments have been made by some in promoting vaccination policies, as well. Generally, vaccine effectiveness cannot be accurately estimated from raw population summaries but instead require rigorous, careful studies using epidemiologic designs and statistical analysis tools attempting to adjust for key confounders and sources of bias. This article summarizes what aggregated evidence across studies reveals about effectiveness of the mRNA vaccines as the pandemic has evolved, chronologically summarized with emerging variants and highlighting some of the fallacies and flawed arguments feeding social media-based claims that have obscured society's collective understanding.
Collapse
Affiliation(s)
- Jeffrey S Morris
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
11
|
Yang W, Parton H, Li W, Watts EA, Lee E, Yuan H. SARS-CoV-2 dynamics in New York City during March 2020-August 2023. COMMUNICATIONS MEDICINE 2025; 5:102. [PMID: 40195487 PMCID: PMC11977191 DOI: 10.1038/s43856-025-00826-6] [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: 08/15/2024] [Accepted: 03/28/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been widespread since 2020 and will likely continue to cause substantial recurring epidemics. However, understanding the underlying infection burden and dynamics, particularly since late 2021 when the Omicron variant emerged, is challenging. Here, we leverage extensive surveillance data available in New York City (NYC) and a comprehensive model-inference system to reconstruct SARS-CoV-2 dynamics therein through August 2023. METHODS We fit a metapopulation network SEIRSV (Susceptible-Exposed-Infectious-(re)Susceptible-Vaccination) model to age- and neighborhood-specific data of COVID-19 cases, emergency department visits, and deaths in NYC from the pandemic onset in March 2020 to August 2023. We further validate the model-inference estimates using independent SARS-CoV-2 wastewater viral load data. RESULTS The validated model-inference estimates indicate a very high infection burden-the number of infections (i.e., including undetected asymptomatic/mild infections) totaled twice the population size ( > 5 times documented case count) during the first 3.5 years. Estimated virus transmissibility increased around 3-fold, whereas estimated infection-fatality risk (IFR) decreased by >10-fold during this period. The detailed estimates also reveal highly complex variant dynamics and immune landscape, and higher infection risk during winter in NYC over the study period. CONCLUSIONS This study provides highly detailed epidemiological estimates and identifies key transmission dynamics and drivers of SARS-CoV-2 during its first 3.5 years of circulation in a large urban center (i.e., NYC). These transmission dynamics and drivers may be relevant to other populations and inform future planning to help mitigate the public health burden of SARS-CoV-2.
Collapse
Affiliation(s)
- Wan Yang
- Department of Epidemiology, Columbia University, New York, NY, USA.
| | - Hilary Parton
- New York City Department of Health and Mental Hygiene, Queens, NY, USA
| | - Wenhui Li
- New York City Department of Health and Mental Hygiene, Queens, NY, USA
| | - Elizabeth A Watts
- New York City Department of Health and Mental Hygiene, Queens, NY, USA
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Ellen Lee
- New York City Department of Health and Mental Hygiene, Queens, NY, USA
| | - Haokun Yuan
- Department of Epidemiology, Columbia University, New York, NY, USA
| |
Collapse
|
12
|
da Costa FADS, Assato PA, Tasca KI, de Moraes LN, Dos Santos Figueiredo DB, Ribeiro G, Lima ARJ, Sampaio SC, Elias Sabbaga MCQB, Fortaleza CMCB, de Souza-Neto JA, Grotto RMT. Genomic and epidemiological surveillance of SARS-CoV-2 variants during the pre-mass vaccination period in Botucatu, Brazil. Virus Genes 2025; 61:153-166. [PMID: 39928246 DOI: 10.1007/s11262-025-02137-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/30/2025] [Indexed: 02/11/2025]
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has caused an unprecedented pandemic in human history. To date, more than 6.5 million lives have been lost to COVID-19 (coronavirus disease 2019). Following widespread efforts for COVID-19 vaccination in Brazil, there has been a drastic reduction in COVID-19 deaths, which was particularly evident in the city of Botucatu, SP, Brazil, after a campaign to a mass vaccination. Our objective was to assess the temporal and epidemiological spread of variants of concern (VOC) of SARS-CoV-2 in the four weeks preceding the massive vaccination campaign in the municipality. After randomizing 400 samples, Next-Generation Sequencing was used to produce sequences and determine the variants. Among high-quality sequences, 98.4% belonged to the VOC Gamma, with P.1 and P.1.14 being the most prevalent lineages. P.1 was more frequent in both men and women, and in younger individuals and adults (0-59 years) compared to P.1.14. There was no correlation between the variants and the presence of comorbidities or between them and moderate to severe clinical cases of COVID-19 or death. However, P.1 was more frequent than P.1.14 in people with mild forms of the disease and in those who exhibited symptoms. In the phylogenetic analyses, a small cluster of 9 P.1.14 samples containing mutations in ORF1a: M584V and A3620V was observed, which had not been found in any Gamma sequences to date. The importance of genomic surveillance of SARS-CoV-2 is evident in assisting public health decision-making and the management of COVID-19 and other diseases.
Collapse
Affiliation(s)
- Felipe Allan da Silva da Costa
- School of Agricultural Sciences, São Paulo State University (Unesp), Botucatu, SP, Brazil
- Clinical Hospital of the Botucatu Medical School, Av. Professor Mário Rubens Guimarães Montenegro, No Number-Jardim São José, Botucatu, SP, 18618-970, Brazil
| | - Patricia Akemi Assato
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Karen Ingrid Tasca
- Medical School, São Paulo State University (Unesp), Botucatu, SP, Brazil
| | - Leonardo Nazário de Moraes
- Clinical Hospital of the Botucatu Medical School, Av. Professor Mário Rubens Guimarães Montenegro, No Number-Jardim São José, Botucatu, SP, 18618-970, Brazil
| | | | | | | | | | | | | | - Jayme Augusto de Souza-Neto
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, USA.
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, 1800 Denison Ave, Manhattan, KS, 66506, USA.
| | - Rejane Maria Tommasini Grotto
- School of Agricultural Sciences, São Paulo State University (Unesp), Botucatu, SP, Brazil.
- Clinical Hospital of the Botucatu Medical School, Av. Professor Mário Rubens Guimarães Montenegro, No Number-Jardim São José, Botucatu, SP, 18618-970, Brazil.
| |
Collapse
|
13
|
Suomenrinne-Nordvik A, Leino T, Shubin M, Auranen K, Vänskä S. Quantifying the direct and indirect components of COVID-19 vaccine effectiveness during the Delta variant era. Epidemiol Infect 2025; 153:e59. [PMID: 40123413 PMCID: PMC12001148 DOI: 10.1017/s0950268825000354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/29/2024] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
The efficacy of COVID-19 vaccines against the Delta variant has been observed to be high, both against severe disease and infection. The full population level vaccine effectiveness, however, also contains the indirect effects of vaccination, which require analysis of transmission dynamics to uncover. Finland was close to naïve to SARS-CoV-2 infections before the Delta dominant era, and non-pharmaceutical interventions (NPIs) were at an internationally low level. We utilize Finnish register data and a mathematical model for transmission and COVID-19 disease burden to construct a completely unvaccinated control population and estimate the different components of the vaccine effectiveness. The estimated direct effectiveness was 72% against COVID-19 cases and 87-96% against severe disease outcomes, but the estimated indirect effectiveness was even better, 93% against cases and 94-97% against severe disease. The total and overall effectiveness, including both direct and indirect effects of vaccination, were thus excellent. Our results show how well the population was protected by vaccination during the Delta era, especially by the indirect effectiveness, providing protection also to the unvaccinated part of the population. The estimated averted numbers of hospitalizations, ICU admissions, and deaths in Finland during the Delta era under the implemented NPIs were about 100 times the observed numbers.
Collapse
Affiliation(s)
| | - Tuija Leino
- Department of Public Health, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Mikhail Shubin
- Department of Public Health, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
| | - Kari Auranen
- Department of Public Health, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
| | - Simopekka Vänskä
- Department of Public Health, Finnish Institute for Health and Welfare, Helsinki, Finland
| |
Collapse
|
14
|
Monge S, Humphreys J, Nicolay N, Braeye T, Van Evercooren I, Hansen CH, Emborg HD, Fabiani M, Sacco C, Castilla J, Martínez-Baz I, de Gier B, Hahné S, Meijerink H, Kristoffersen AB, Machado A, Soares P, Fontán-Vela M, Nardone A, Kissling E, Nunes B, VEBIS-Lot 4 working group. Comparison of two methods for the estimation of COVID-19 vaccine effectiveness of the autumnal booster within the VEBIS-EHR network in 2022/23. Epidemiol Infect 2025; 153:e54. [PMID: 40090768 PMCID: PMC12001144 DOI: 10.1017/s0950268825000317] [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: 04/01/2024] [Revised: 12/09/2024] [Accepted: 03/06/2025] [Indexed: 03/18/2025] Open
Abstract
Within an infrastructure to monitor vaccine effectiveness (VE) against hospitalization due to COVID-19 and COVID-19 related deaths from November 2022 to July 2023 in seven countries in real-world conditions (VEBIS network), we compared two approaches: (a) estimating VE of the first, second or third COVID-19 booster doses administered during the autumn of 2022, and (b) estimating VE of the autumn vaccination dose regardless of the number of prior doses (autumnal booster approach). Retrospective cohorts were constructed using Electronic Health Records at each participating site. Cox regressions with time-changing vaccination status were fit and site-specific estimates were combined using random-effects meta-analysis. VE estimates with both approaches were mostly similar, particularly shortly after the start of the vaccination campaign, and showed a similar timing of VE waning. However, autumnal booster estimates were more precise and showed a clearer trend, particularly compared to third booster estimates, as calendar time increased after the vaccination campaign and during periods of lower SARS-CoV-2 activity. Moreover, the decrease in protection by increasing calendar time was more clear and precise than when comparing protection by number of doses. Therefore, estimating VE under an autumnal booster framework emerges as a preferred method for future monitoring of COVID-19 vaccination campaigns.
Collapse
Affiliation(s)
- Susana Monge
- Department of Communicable Diseases, National Centre of Epidemiology, Institute of Health Carlos III, Madrid, Spain
- CIBER on Infectious Diseases, Madrid, Spain
| | | | - Nathalie Nicolay
- Vaccine Preventable Diseases and Immunisation, European Centre for Disease Prevention and Control (ECDC), Solna, Sweden
| | - Toon Braeye
- Department of Epidemiology and Public Health, Sciensano, Elsene, Belgium
| | | | - Christian Holm Hansen
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
| | - Hanne-Dorthe Emborg
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
| | - Massimo Fabiani
- Infectious Diseases Department, Istituto Superiore di Sanità, Rome, Italy
| | - Chiara Sacco
- Infectious Diseases Department, Istituto Superiore di Sanità, Rome, Italy
- European Programme on Intervention Epidemiology Training (EPIET), European Centre for Disease Prevention and Control, Stockholm, Sweden
| | - Jesús Castilla
- Instituto de Salud Pública de Navarra – IdiSNA, Pamplona, Spain
- CIBER on Epidemiology and Public Health, Madrid, Spain
| | - Iván Martínez-Baz
- Instituto de Salud Pública de Navarra – IdiSNA, Pamplona, Spain
- CIBER on Epidemiology and Public Health, Madrid, Spain
| | - Brechje de Gier
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Susan Hahné
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Hinta Meijerink
- Department of Infection Control and Vaccines, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Anja Bråthen Kristoffersen
- Department of Method Development and Analytics, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Ausenda Machado
- Departamento de Epidemiologia, Instituto Nacional de Saúde Doutor Ricardo Jorge, LisboaPortugal
| | - Patricia Soares
- Departamento de Epidemiologia, Instituto Nacional de Saúde Doutor Ricardo Jorge, LisboaPortugal
| | - Mario Fontán-Vela
- Department of Communicable Diseases, National Centre of Epidemiology, Institute of Health Carlos III, Madrid, Spain
- Public Health and Epidemiology Research Group, School of Medicine and Health Sciences, Universidad de Alcalá, Alcalá de Henares, Spain
| | | | | | | | | |
Collapse
|
15
|
Ferrante L, Capanema E, Steinmetz WAC, Nelson BW, Almeida ACL, Leão J, Pereira LS, Vassão RC, Fearnside PM, Tupinambás U. High transmission of SARS-CoV-2 in Amazonia, Brazil: an epidemiological strategy to contain severe cases of COVID-19. J Public Health Policy 2025; 46:71-86. [PMID: 39638867 DOI: 10.1057/s41271-024-00530-0] [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] [Accepted: 10/22/2024] [Indexed: 12/07/2024]
Abstract
We followed the COVID-19 pandemic in Manaus, one of the epicenters of COVID-19 in Brazil, using an epidemiological mathematical model and made five main conclusions. First, in early 2022, the actual cases exceed officially reported data by up to 8 times. Second, despite vaccination campaigns, the collective immunity threshold necessary was insufficient to contain severe cases of COVID-19. Next, the low observed mortality demonstrated the effectiveness of vaccination. Next, the drop in the vaccination rate combined with immune escape by the Omicron sub-variants (BA.2.12.1, BA.4, and BA.5) resulted in new wave after November 2022. Finally, to minimize severe cases of COVID-19, we need to raise vaccination thresholds above 90-95% of the entire population including children aged 6 months and older and require booster doses at least in four-month intervals. This approach would help to prevent severe cases of COVID-19 that cause hospitalizations and deaths.
Collapse
Affiliation(s)
- Lucas Ferrante
- School of Arts, Sciences, and Humanities - (EACH), University of São Paulo (USP), São Paulo, São Paulo, Brazil.
- Laboratory of Animal Genetics and Evolution (LEGAL), Federal University of Amazonas (UFAM), Manaus, Amazonas, Brazil.
| | - Eduardo Capanema
- Computational Mathematics Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | | | - Bruce Walker Nelson
- Department of Environmental Dynamics, National Institute for Amazonian Research (INPA), Manaus, Amazonas, Brazil
| | | | - Jeremias Leão
- Department of Statistics, Federal University of Amazonas (UFAM), Manaus, Amazonas, Brazil
| | | | | | - Philip Martin Fearnside
- Department of Environmental Dynamics, National Institute for Amazonian Research (INPA), Manaus, Amazonas, Brazil
| | - Unaí Tupinambás
- Department of Internal Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
16
|
Silva Ramírez B, Peñuelas Urquides K, Escobedo Guajardo BL, Mata Tijerina VL, Cruz Luna JE, Corrales Pérez R, Gómez García S, González Escalante LA, Camacho Moll ME. Assessment of COVID-19 Vaccine Effectiveness Against SARS-CoV-2 Infection, Hospitalization and Death in Mexican Patients with Metabolic Syndrome from Northeast Mexico: A Multicenter Study. Vaccines (Basel) 2025; 13:244. [PMID: 40266114 PMCID: PMC11945729 DOI: 10.3390/vaccines13030244] [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: 11/25/2024] [Revised: 12/27/2024] [Accepted: 01/01/2025] [Indexed: 04/24/2025] Open
Abstract
Background/Objectives: Metabolic syndrome (MetS) is a predisposing factor for severe COVID-19. The effectiveness of COVID-19 vaccines in patients with MetS has been poorly investigated. The aim of this study was to evaluate the effectiveness of COVID-19 vaccination before (BO) and after the Omicron (AO) SARS-CoV-2 variant in patients with MetS. Methods: This retrospective observational study was carried out in a total of 3194 patients with MetS and a COVID-19 PCR or rapid antigen test. The main outcomes were vaccine effectiveness against infection, hospitalization and death resulting from COVID-19. Results: BO, only two doses of BNT162b2 were effective against infection, this effectiveness was lost AO. Also, with two doses, BNT162b2, ChAdOx1 and CoronaVac were effective against hospitalization BO; however, AO, only BNT162b2 and CoronaVac were effective. Regarding death as an outcome of COVID-19, two doses of BNT162b2 were effective BO, whereas AO, BNT162b2 and CoronaVac were 100% effective. BO the presentation of a sore throat increased after two doses of COVID-19 vaccine regardless of the type, and the presentation of dyspnea diminished after two doses of BNT162b2 and CoronaVac. Conclusions: The SARS-CoV-2 Omicron variant has impacted vaccines' effectiveness against hospitalization and death in patients with MetS. A tailored vaccination scheme for patients with MetS should be implemented due to the varying effectiveness rates observed in our study.
Collapse
Affiliation(s)
- Beatriz Silva Ramírez
- Laboratory of Immunogenetics, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Nuevo Leon, Mexico; (B.S.R.); (V.L.M.T.)
| | - Katia Peñuelas Urquides
- Laboratory of Molecular Microbiology, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Nuevo Leon, Mexico; (K.P.U.); (L.A.G.E.)
| | - Brenda Leticia Escobedo Guajardo
- Laboratory of Molecular Research of Diseases, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Nuevo Leon, Mexico;
| | - Viviana Leticia Mata Tijerina
- Laboratory of Immunogenetics, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Nuevo Leon, Mexico; (B.S.R.); (V.L.M.T.)
| | - Jorge Eleazar Cruz Luna
- Medical Epidemiological Assistance Coordination of the State of Nuevo Leon, Mexican Social Security Institute, Monterrey 64000, Nuevo Leon, Mexico; (J.E.C.L.); (R.C.P.); (S.G.G.)
| | - Roberto Corrales Pérez
- Medical Epidemiological Assistance Coordination of the State of Nuevo Leon, Mexican Social Security Institute, Monterrey 64000, Nuevo Leon, Mexico; (J.E.C.L.); (R.C.P.); (S.G.G.)
| | - Salvador Gómez García
- Medical Epidemiological Assistance Coordination of the State of Nuevo Leon, Mexican Social Security Institute, Monterrey 64000, Nuevo Leon, Mexico; (J.E.C.L.); (R.C.P.); (S.G.G.)
| | - Laura Adiene González Escalante
- Laboratory of Molecular Microbiology, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Nuevo Leon, Mexico; (K.P.U.); (L.A.G.E.)
| | - María Elena Camacho Moll
- Laboratory of Molecular Biology, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Nuevo Leon, Mexico
| |
Collapse
|
17
|
Yang H, Li J, Song C, Li H, Luo Q, Chen M. Emerging Gene Therapy Based on Nanocarriers: A Promising Therapeutic Alternative for Cardiovascular Diseases and a Novel Strategy in Valvular Heart Disease. Int J Mol Sci 2025; 26:1743. [PMID: 40004206 PMCID: PMC11855571 DOI: 10.3390/ijms26041743] [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: 12/31/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Cardiovascular disease remains a leading cause of global mortality, with many unresolved issues in current clinical treatment strategies despite years of extensive research. Due to the great progress in nanotechnology and gene therapy in recent years, the emerging gene therapy based on nanocarriers has provided a promising therapeutic alternative for cardiovascular diseases. This review outlines the status of nanocarriers as vectors in gene therapy for cardiovascular diseases, including coronary heart disease, pulmonary hypertension, hypertension, and valvular heart disease. It discusses challenges and future prospects, aiming to support emerging clinical treatments. This review is the first to summarize gene therapy using nanocarriers for valvular heart disease, highlighting their potential in targeting challenging tissues.
Collapse
Affiliation(s)
- Haoran Yang
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Junli Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chengxiang Song
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Hongde Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Qiang Luo
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mao Chen
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
18
|
Chen J, Lei Y, Wu Q, Zhou T, Zhang B, Becich MJ, Bisyuk Y, Blecker S, Chrischilles EA, Christakis DA, Cowell LG, Cummins MR, Fernandez SA, Fort D, Gonzalez S, Herring SJ, Horne BD, Horowitz C, Liu M, Kim S, Mirhaji P, Mosa ASM, Muszynski JA, Paules CI, Sato A, Schwenk HT, Sengupta S, Suresh S, Taylor BW, Williams DA, He Y, Morris JS, Jhaveri R, Forrest CB, Chen Y. Vaccine Effectiveness Among 5- to 17-year-old Individuals with Prior SARS-CoV-2 Infection: An EHR-Based Target Trial Emulation Study from the RECOVER Project. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.07.25321814. [PMID: 39974088 PMCID: PMC11838676 DOI: 10.1101/2025.02.07.25321814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
IMPORTANCE Prior studies have demonstrated the effectiveness of COVID-19 vaccines in children and adolescents. However, the benefits of vaccination in these age groups with prior infection remain underexplored. OBJECTIVE To evaluate the effectiveness of COVID-19 vaccination in preventing reinfection with various Omicron subvariants (BA.1/2, BA.4/5, XBB, and later) among 5- to 17-year-olds with prior SARS-CoV-2 infection. DESIGN A target trial emulation through nested designs with distinct study periods. SETTING The study utilized data from the Research COVID to Enhance Recovery (RECOVER) initiative, a national electronic health record (EHR) database comprising 37 U.S. children's hospitals and health institutions. PARTICIPANTS Individuals aged 5-17 years with a documented history of SARS-CoV-2 infection prior to the study start date during a specific variant-dominant period (Delta, BA.1/2, or BA.4/5) who received a subsequent dose of COVID-19 vaccine during the study periods were compared with those with a documented history of infection who did not receive SARS-CoV-2 vaccine during the study period. Those infected within the Delta-Omicron composite period (December 1, 2021, to December 31, 2021) were excluded. The study period was from January 1, 2022, to August 30, 2023, and focused on adolescents aged 12 to 17 years and children aged 5 to 11 years. EXPOSURES At least received one COVID-19 vaccination during the study period vs. no receipt of any COVID-19 vaccine during the study period. MAIN OUTCOMES AND MEASURES The primary outcome is documented SARS-CoV-2 reinfection during the study period (both asymptomatic and symptomatic cases). The effectiveness of the COVID-19 vaccine was estimated as (1- hazard ratio) *100%, with confounders adjusted by a combination of propensity score matching and exact matching. RESULTS The study analyzed 87,573 participants during the BA.1/2 period, 229,326 during the BA.4/5 period, and 282,981 during the XBB or later period. Among vaccinated individuals, significant protection was observed during the BA.1/2 period, with effectiveness rates of 62% (95% CI: 38%-77%) for children and 65% (95% CI: 32%-81%) for adolescents. During the BA.4/5 period, vaccine effectiveness was 57% (95% CI: 25%-76%) for children, but not statistically significant for adolescents (36%, 95% CI: -16%-65%). For the XBB period, no significant protection was observed in either group, with effectiveness rates of 22% (95% CI: -36%-56%) in children and 34% (95% CI: -10%-61%) in adolescents. CONCLUSIONS AND RELEVANCE COVID-19 vaccination provides significant protection against reinfection for children and adolescents with prior infections during the early and mid-Omicron periods. This study also highlights the importance of addressing low vaccination rates in pediatric populations to enhance protection against emerging variants.
Collapse
Affiliation(s)
- Jiajie Chen
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yuqing Lei
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Qiong Wu
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Biostatistics and Health Data Science, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ting Zhou
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bingyu Zhang
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia, PA, USA
- The Graduate Group in Applied Mathematics and Computational Science, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael J. Becich
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburg, PA, USA
| | - Yuriy Bisyuk
- University Medical Center New Orleans, New Orleans, LA, USA
| | - Saul Blecker
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Dimitri A. Christakis
- Center for Child Health, Behavior and Development, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Lindsay G. Cowell
- Peter O’Donnell Jr. School of Public Health, Department of Immunology, School of Biomedical Sciences, UT Southwestern Medical Center; Dallas, TX, USA
| | | | - Soledad A. Fernandez
- Department of Biomedical Informatics and Center for Biostatistics, Ohio State University, Columbus, OH, USA
| | - Daniel Fort
- Center for Outcomes Research, Ochsner Health, New Orleans, LA, USA
| | | | - Sharon J. Herring
- Program for Maternal Health Equity, Center for Urban Bioethics, Department of Population Health and Urban Bioethics, Center for Obesity Research and Education, College of Public Health, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Benjamin D. Horne
- Intermountain Medical Center Heart Institute, Salt Lake City, UT, USA
| | - Carol Horowitz
- Institute for Health Equity Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mei Liu
- Department of Health Outcomes and Biomedical Informatics, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Susan Kim
- University of California, San Francisco, Division of Rheumatology, Benioff Children’s Hospital, San Francisco, CA, USA
| | | | - Abu Saleh Mohammad Mosa
- Department of Biomedical Informatics, Biostatistics, and Medical Epidemiology, University of Missouri School of Medicine, Columbia, MO, USA
| | - Jennifer A. Muszynski
- Division of Critical Care Medicine, Department of Pediatrics, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Catharine I. Paules
- Division of Infectious Diseases, Department of Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Alice Sato
- Division of Pediatric Infectious Diseases, University of Nebraska Medical Center, and Children’s Hospital & Medical Center, Omaha, NE, USA
| | - Hayden T. Schwenk
- Stanford School of Medicine, Division of Pediatric Infectious Diseases, Stanford, CA, USA
| | - Soumitra Sengupta
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Srinivasan Suresh
- Department of Pediatrics, University of Pittsburgh, and UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Bradley W. Taylor
- Clinical and Translational Science Institute, The Medical College of Wisconsin, Milwaukee, WI, USA
| | - David A. Williams
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Yongqun He
- Medical School, University of Michigan, Ann Arbor, Michigan, USA
| | - Jeffrey S. Morris
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ravi Jhaveri
- Division of Pediatric Infectious Diseases, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Christopher B Forrest
- Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
- Applied Clinical Research Center, Children’s Hospital of Philadelphia, Department of Healthcare Management, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, USA
| | - Yong Chen
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- The Graduate Group in Applied Mathematics and Computational Science, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Penn Medicine Center for Evidence-based Practice (CEP), Philadelphia, PA, USA
- Penn Institute for Biomedical Informatics (IBI), Philadelphia, PA, USA
| |
Collapse
|
19
|
Shintani Y, Yamamoto H, Sato Y, Inoue M, Asakura K, Ito H, Uramoto H, Okada Y, Sato T, Fukui M, Hoshikawa Y, Chen-Yoshikawa TF, Chida M, Ikeda N, Yoshino I. Effects of the COVID-19 pandemic on surgical treatment for thoracic malignant tumor cases in Japan: a national clinical database analysis. Surg Today 2025; 55:265-272. [PMID: 39644388 PMCID: PMC11757856 DOI: 10.1007/s00595-024-02907-w] [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: 04/03/2024] [Accepted: 07/15/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVE Surgical care has been significantly affected by the COVID-19 pandemic. This study was conducted to evaluate the effects of the pandemic on lung cancer and mediastinal tumor surgery. METHODS Changes in the number of surgical procedures for lung cancer and mediastinal tumors were analyzed using the National Clinical Database of Japan. Patient characteristics, including disease stage and histological type, from 2019 to 2022 were evaluated using annual datasets. RESULTS Comparisons with 2019 showed that the number of patients who underwent surgery for primary lung cancer or a mediastinal tumor decreased in 2020 and then remained stable. There were no clinically significant changes in the trend over the four-year period regarding the number of patients for each clinical and pathological stage of lung cancer. Regarding mediastinal tumors, there was no significant difference in tumor size between years. There was a slight change in the selection of surgical indication during the second quarter of 2020, although its impact on annual trends in the stage distribution for lung cancer and primary disease for mediastinal tumors was minimal. CONCLUSIONS Analyses of lung cancer and mediastinal tumor surgery cases in Japan during the COVID-19 pandemic showed no significant disease profile changes related to treatment delay.
Collapse
Affiliation(s)
- Yasushi Shintani
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, 2-2-L5, Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hiroyuki Yamamoto
- Department of Healthcare Quality Assessment, The University of Tokyo, Tokyo, Japan
| | - Yukio Sato
- Department of Thoracic Surgery, University of Tsukuba, Ibaraki, Japan
| | - Masayoshi Inoue
- Division of Thoracic Surgery, Department Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Keisuke Asakura
- Division of Thoracic Surgery, Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Ito
- Department of Thoracic Surgery, Kanagawa Cancer Center, Kanagawa, Japan
| | - Hidetaka Uramoto
- Department of Thoracic Surgery, Kanazawa Medical University, Ishikawa, Japan
| | - Yoshinori Okada
- Department of Thoracic Surgery, Tohoku University, Miyagi, Japan
| | - Toshihiko Sato
- Department of General Thoracic, Breast and Pediatric Surgery, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Mariko Fukui
- Department of General Thoracic Surgery, Juntendo University, Tokyo, Japan
| | - Yasushi Hoshikawa
- Department of Thoracic Surgery, Fujita Health University School of Medicine, Aichi, Japan
| | | | - Masayuki Chida
- Department of General Thoracic Surgery, Dokkyo Medical University, Tochigi, Japan
| | - Norihiko Ikeda
- Department of Surgery, Tokyo Medical University, Tokyo, Japan
| | - Ichiro Yoshino
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Thoracic Surgery, International University of Health and Welfare School of Medicine, Chiba, Japan
| |
Collapse
|
20
|
González M, Al-Adib M, Rodríguez AB, Carrasco C. Factors associated with menstrual-related disturbances following SARS-CoV-2 vaccination: a Spanish retrospective observational study in formerly menstruating women. Women Health 2025; 65:167-181. [PMID: 39819300 DOI: 10.1080/03630242.2025.2451360] [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: 03/01/2024] [Revised: 12/12/2024] [Accepted: 01/05/2025] [Indexed: 01/19/2025]
Abstract
BACKGROUND A growing body of evidence suggests a potential link between the SARS-CoV-2 vaccine and menstrual changes in women who were menstruating at the time of vaccination. Nevertheless, the prevalence of this event in those with secondary amenorrhea for different causes, i.e. formerly menstruating women, remains unclear. It is plausible that, analogous to those observed in currently menstruating women, they experienced some degree of alteration in their reproductive health, defined here as menstrual-related disturbances. OBJECTIVE The aim was to analyze this phenomenon and identify the factors associated with the occurrence of menstrual-related disturbances in this subpopulation. Study design: A retrospective observational cross-sectional study was conducted among adult Spanish in December 2021 using an online survey (N = 17,512). The present analysis includes a subpopulation of vaccinated and formerly menstruating women (N = 548). General characteristics, medical history, and adverse events following COVID-19 vaccination were recorded. Chi-square, Mann-Whitney U and McNemar mid-P tests were performed. Bivariate logistic regression was then used to identify the key factors influencing this unexpected event. RESULTS In comparison with the first dose, significantly higher percentages of respondents experienced menstrual-related disturbances (dose 1: 38.5 percent vs. dose 2: 44.8 percent) after receiving the second one. Among them, those related to the length and flow stand out, being of long-term nature in about 17-20 percent of cases. Interindividual factors influencing this unexpected event after receiving the dose 1 may include weight, perimenopause, preexisting diagnoses of non-autoimmune rheumatic/articular conditions, use of hormonal contraceptives, suffering from other vaccine side effects - such as arm pain and the number of previous pregnancies; for dose 2, these factors may include suffering from menstrual-related alterations after receiving dose 1, as well as the use of hormonal contraceptives and perimenopause. CONCLUSION Formerly menstruating women might experience long-term menstrual-related disturbances following COVID-19 vaccination. Potential influencing factors include weight, perimenopause, rheumatic/articular conditions, hormonal contraceptives, vaccine side effects and previous pregnancies.
Collapse
Affiliation(s)
- María González
- Neuroimmunophysiology and Chrononutrition Research Group, Department of Physiology, Universidad de Extremadura, Badajoz, Spain
| | - Miriam Al-Adib
- Neuroimmunophysiology and Chrononutrition Research Group, Department of Physiology, Universidad de Extremadura, Badajoz, Spain
- Obstetrics & Gynecology clinics "Miriam Gine", Badajoz, Spain
| | - Ana B Rodríguez
- Neuroimmunophysiology and Chrononutrition Research Group, Department of Physiology, Universidad de Extremadura, Badajoz, Spain
| | - Cristina Carrasco
- Neuroimmunophysiology and Chrononutrition Research Group, Department of Physiology, Universidad de Extremadura, Badajoz, Spain
| |
Collapse
|
21
|
Sultana A, Banu LA, Hossain M, Azmin N, Nila NN, Sinha SK, Hassan Z. Evaluation of Genomic Surveillance of SARS-CoV-2 Virus Isolates and Comparison of Mutational Spectrum of Variants in Bangladesh. Viruses 2025; 17:182. [PMID: 40006937 PMCID: PMC11860708 DOI: 10.3390/v17020182] [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: 11/28/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 02/27/2025] Open
Abstract
The SARS-CoV-2-induced disease, COVID-19, remains a worldwide public health concern due to its high rate of transmission, even in vaccinated and previously infected people. In the endemic state, it continues to cause significant pathology. To elu- cidate the viral mutational changes and screen the emergence of new variants of concern, we conducted this study in Bangladesh. The viral RNA genomes extracted from 25 ran- domly collected samples of COVID-19-positive patients from March 2021 to February 2022 were sequenced using Illumina COVID Seq protocol and genomic data processing, as well as evaluations performed in DRAGEN COVID Lineage software. In this study, the percentage of Delta, Omicron, and Mauritius variants identified were 88%, 8%, and 4%, respectively. All of the 25 samples had 23,403 A>G (D614G, S gene), 3037 C>T (nsp3), and 14,408 C>T (nsp12) mutations, where 23,403 A>G was responsible for increased transmis- sion. Omicron had the highest number of unique mutations in the spike protein (i.e., sub- stitutions, deletions, and insertions), which may explain its higher transmissibility and immune-evading ability than Delta. A total of 779 mutations were identified, where 691 substitutions, 85 deletions, and 3 insertion mutations were observed. To sum up, our study will enrich the genomic database of SARS-CoV-2, aiding in treatment strategies along with understanding the virus's preferences in both mutation type and mutation site for predicting newly emerged viruses' survival strategies and thus for preparing to coun- teract them.
Collapse
Affiliation(s)
- Abeda Sultana
- Department of Anatomy, Dhaka Medical College, Dhaka 1000, Bangladesh;
| | - Laila Anjuman Banu
- Department of Anatomy, Dhaka Medical College, Dhaka 1000, Bangladesh;
- Genetics and Molecular Biology Laboratory, Bangabandhu Sheikh Mujib Medical University, Dhaka 1000, Bangladesh
| | - Mahmud Hossain
- Laboratory of Neuroscience and Neurogenetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh; (M.H.); (N.N.N.)
| | - Nahid Azmin
- Department of Anatomy, Shahabuddin Medical College, Dhaka 1212, Bangladesh;
| | - Nurun Nahar Nila
- Laboratory of Neuroscience and Neurogenetics, Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh; (M.H.); (N.N.N.)
| | - Sharadindu Kanti Sinha
- Department of Pharmacology, Bangabandhu Sheikh Mujib Medical University, Dhaka 1000, Bangladesh;
| | - Zahid Hassan
- Department of Physiology and Molecular Biology, Bangladesh University of Health Sciences, Dhaka 1216, Bangladesh;
| |
Collapse
|
22
|
Arghal R, Rubin H, Saeedi Bidokhti S, Sarkar S. Protect or prevent? A practicable framework for the dilemmas of COVID-19 vaccine prioritization. PLoS One 2025; 20:e0316294. [PMID: 39841676 PMCID: PMC11753641 DOI: 10.1371/journal.pone.0316294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
Determining COVID-19 vaccination strategies presents many challenges in light of limited vaccination capacity and the heterogeneity of affected communities. Who should be prioritized for early vaccination when different groups manifest different levels of risks and contact rates? Answering such questions often becomes computationally intractable given that network size can exceed millions. We obtain a framework to compute the optimal vaccination strategy within seconds to minutes from among all strategies, including highly dynamic ones that adjust vaccine allocation as often as required, and even with modest computation resources. We then determine the optimal strategy for a large range of parameter values representative of various US states, countries, and case studies including retirement homes and prisons. The optimal is almost always one of a few candidate strategies, and, even when not, the suboptimality of the best among these candidates is minimal. Further, we find that many commonly deployed vaccination strategies, such as vaccinating the high risk group first, or administering second doses without delay, can often incur higher death rates, hospitalizations, and symptomatic infection counts. Our framework can be easily adapted to future variants or pandemics through appropriate choice of the compartments of the disease and parameters.
Collapse
Affiliation(s)
- Raghu Arghal
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Harvey Rubin
- Division of Infectious Diseases, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Shirin Saeedi Bidokhti
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Saswati Sarkar
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA, United States of America
| |
Collapse
|
23
|
Kirsebom FCM, Hall V, Stowe J. How do large-scale population studies inform vaccine evaluations in England? Clin Exp Immunol 2025; 219:uxaf006. [PMID: 39910973 PMCID: PMC11933692 DOI: 10.1093/cei/uxaf006] [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: 11/05/2024] [Revised: 12/16/2024] [Accepted: 02/04/2025] [Indexed: 02/07/2025] Open
Abstract
Large-scale population studies are important to monitor and evaluate aspects of a vaccination programme including vaccine coverage, real-world effectiveness, and post-licensure vaccine safety. These types of epidemiological studies often come under the remit of public health agencies, such as the UK Health Security Agency (UKHSA) in England, which are required to undertake surveillance of vaccine-preventable diseases, including via seroepidemiological studies and data linkage studies using national-level electronic healthcare data. An individual-level national vaccine register with an accurate denominator can be the key to gaining insights into vaccine coverage, effectiveness, and safety. During the coronavirus disease 2019 pandemic, England's first vaccine register was developed. This enabled timely estimates of real-world vaccine effectiveness in the whole population of England, as well as enabling epidemiological investigations of rare potential risks from vaccines in specific populations. Population-based research studies, including prospective cohort studies, are complementary to surveillance and combined, enable more comprehensive assessments. As there was an unprecedented investment into research studies and infrastructure during the pandemic, the scale of these studies meant they were able to contribute to vaccine programme evaluations in a way that had not been possible for previous vaccine programmes. In this review, we summarise the different large-scale surveillance and research studies that have been used to evaluate and inform vaccine policy from the time of the first data linkage studies undertaken in England in the 1990s to the present-day post-COVID-19 pandemic.
Collapse
Affiliation(s)
- Freja C M Kirsebom
- Immunisation and Vaccine Preventable Diseases Department, UK Health Security Agency, London, UK
| | | | - Julia Stowe
- Immunisation and Vaccine Preventable Diseases Department, UK Health Security Agency, London, UK
| |
Collapse
|
24
|
Meng X, Fan Y, Qiao Y, Lin J, Cai Z, Si S. Evolutionary analysis of a coupled epidemic-voluntary vaccination behavior model with immunity waning on complex networks. RISK ANALYSIS : AN OFFICIAL PUBLICATION OF THE SOCIETY FOR RISK ANALYSIS 2025. [PMID: 39826914 DOI: 10.1111/risa.17699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 11/11/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025]
Abstract
Vaccination is the most effective method of preventing and controlling the transmission of infectious diseases within populations. However, the phenomenon of waning immunity can induce periodic fluctuations in epidemic spreading. This study proposes a coupled epidemic-vaccination dynamic model to analyze the influence of immunity waning on the epidemic spreading within the context of voluntary vaccination. First, we establish an SIRSV (susceptible-infected-recovered-susceptible-vaccinated) compartment model to describe the transmission mechanism of infectious diseases based on the mean-field theory. Within this model, we incorporate a nonlinear infection rate with network topology and consider the waning natural and vaccine-induced immunity at the individual level. The evolutionary model of voluntary vaccination strategy is integrated into the SIRSV model to characterize the impact of vaccination behavior on the infectious disease transmission. We also consider two individual risk assessment methods, namely, the individual-based risk assessment (IB-RA) method and the society-based risk assessment (SB-RA) method, originating from local and global perspectives, respectively. Then, utilizing the next-generation matrix method, we derive the time-varying effective reproduction numbers of the model. Also, the theoretical analysis of optimal strategy thresholds in the individual decision-making process is also conducted. The results indicate that the thresholds obtained from the agent-based model (ABM) simulation method are consistent with the theoretical analysis, demonstrating the effectiveness of our model. Finally, we apply the coupled model to the COVID-19 pandemic in France, Germany, Italy, and the United Kingdom. This study analyzes the impact of waning immunity and provides early warning for the outbreak of the epidemics.
Collapse
Affiliation(s)
- Xueyu Meng
- Department of Industrial Engineering, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an, China
- Ministry of Industry and Information Technology Key Laboratory of Industrial Engineering and Intelligent Manufacturing, Northwestern Polytechnical University, Xi'an, China
- Department of Physics, University of Fribourg, Fribourg, Switzerland
| | - Yufei Fan
- Department of Industrial Engineering, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an, China
- Ministry of Industry and Information Technology Key Laboratory of Industrial Engineering and Intelligent Manufacturing, Northwestern Polytechnical University, Xi'an, China
| | - Yanan Qiao
- Department of Physics, University of Fribourg, Fribourg, Switzerland
- State Key Laboratory for Manufacturing Systems Engineering, School of Management, Xian Jiaotong University, Xian, China
| | - Jianhong Lin
- Blockchain & Distributed Ledger Technologies Group, Department of Informatics, University of Zurich, Zurich, Switzerland
- UZH Blockchain Center, University of Zurich, Zurich, Switzerland
| | - Zhiqiang Cai
- Department of Industrial Engineering, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an, China
- Ministry of Industry and Information Technology Key Laboratory of Industrial Engineering and Intelligent Manufacturing, Northwestern Polytechnical University, Xi'an, China
| | - Shubin Si
- Department of Industrial Engineering, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an, China
- Ministry of Industry and Information Technology Key Laboratory of Industrial Engineering and Intelligent Manufacturing, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
25
|
Flores D, Luna EM. The effectiveness of vaccines against COVID-19 in Mexico: A time series approach. Vaccine 2025; 44:126565. [PMID: 39615344 DOI: 10.1016/j.vaccine.2024.126565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 11/12/2024] [Accepted: 11/23/2024] [Indexed: 12/20/2024]
Abstract
OBJECTIVE To estimate the effectiveness of several vaccine brands-Pfizer, Astra, and Sinovac-against symptomatic COVID-19 without information on vaccination at the individual level. METHODS We use data of mass vaccination programs-specifically, for sexagenarians and quinquagenarians-in three large municipalities of Mexico (Monterrey, Guadalupe, and San Nicolás) to conduct a two-step time series estimation procedure involving a synthetic control group. The data covers the period between the first week of March 2020 and the first week of October 2021. RESULTS Vaccine effectiveness is a concave function of time. At the peak, Pfizer reaches 92.6 % effectiveness, Astra 83.6 % and Sinovac 65.6 %. This occurs 9 to 12 weeks after the first shot. CONCLUSION The results indicate that the three vaccines protect against symptomatic COVID-19. Nevertheless, they offer different levels of protection. The results also suggest that VE-under a two-shot scheme-reaches its peak 9 to 16 weeks after the first shot. Moreover, there seems to be a trade-off between achieving higher efficiency by administering the 2nd shot earlier or extending the protection period by administering it later.
Collapse
Affiliation(s)
- D Flores
- Universidad Autónoma de Nuevo León, Facultad de Economía, Campus Mederos, Ave. Lázaro Cárdenas 4600 Ote. Fraccionamiento Las Torres, 64930 Monterrey, NL, Mexico.
| | - E M Luna
- Universidad Autónoma de Nuevo León, Facultad de Economía, Campus Mederos, Ave. Lázaro Cárdenas 4600 Ote. Fraccionamiento Las Torres, 64930 Monterrey, NL, Mexico
| |
Collapse
|
26
|
Brown J, Mook P, Vanhaverbeke M, Gimma A, Hagan J, Singini I, Avdičová M, Cullen G, Dotsenko L, Mossong J, Sadkowska-Todys M, Suija H, Bundle N, Pebody R. Case-only analysis of routine surveillance data: detection of increased vaccine breakthrough infections with SARS-CoV-2 variants in Europe. Epidemiol Infect 2025; 153:e16. [PMID: 39757926 PMCID: PMC11748014 DOI: 10.1017/s0950268824001833] [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: 07/05/2024] [Revised: 11/11/2024] [Accepted: 12/13/2024] [Indexed: 01/07/2025] Open
Abstract
With the ongoing emergence of SARS-CoV-2 variants, there is a need for standard approaches to characterize the risk of vaccine breakthrough. We aimed to estimate the association between variant and vaccination status in case-only surveillance data. Included cases were symptomatic adult laboratory-confirmed COVID-19 cases, with onset between January 2021 and April 2022, reported by five European countries (Estonia, Ireland, Luxembourg, Poland, and Slovakia) to The European Surveillance System. Associations between variant and vaccination status were estimated using conditional logistic regression, within strata of country and calendar date, and adjusting for age and sex. We included 80,143 cases including 20,244 Alpha (B.1.1.7), 152 Beta (B.1.351), 39,900 Delta (B.1.617.2), 361 Gamma (P.1), 10,014 Omicron BA.1, and 9,472 Omicron BA.2. Partially vaccinated cases were more likely than unvaccinated cases to be Beta than Alpha (adjusted odds ratio [aOR] 2.48, 95% CI 1.29-4.74), and Delta than Alpha (aOR 1.75, 1.31-2.34). Fully vaccinated cases were relative to unvaccinated cases more frequently Beta than Alpha (aOR 4.61, 1.89-11.21), Delta than Alpha (aOR 2.30, 1.55-3.39), and Omicron BA.1 than Delta (aOR 1.91, 1.60-2.28). We found signals of increased breakthrough infections for Delta and Beta relative to Alpha, and Omicron BA.1 relative to Delta.
Collapse
Affiliation(s)
- Jeremy Brown
- World Health Organization (WHO) Regional Office for Europe, Copenhagen, Denmark
| | - Piers Mook
- World Health Organization (WHO) Regional Office for Europe, Copenhagen, Denmark
| | | | - Amy Gimma
- World Health Organization (WHO) Regional Office for Europe, Copenhagen, Denmark
| | - José Hagan
- World Health Organization (WHO) Regional Office for Europe, Copenhagen, Denmark
| | - Isaac Singini
- World Health Organization (WHO) Regional Office for Europe, Copenhagen, Denmark
| | - Mária Avdičová
- The Regional Authority of Public Health in Banska Bystrica, Public Health Authority of the Slovak Republic
| | - Gillian Cullen
- Health Protection Surveillance Centre, Dublin, Republic of Ireland
| | | | | | | | | | - Nick Bundle
- European Centre for Disease Prevention and Control (ECDC), Stockholm, Sweden
| | - Richard Pebody
- World Health Organization (WHO) Regional Office for Europe, Copenhagen, Denmark
| |
Collapse
|
27
|
Johnston MD, Pell B, Pemberton J, Rubel DA. The Effect of Vaccination on the Competitive Advantage of Two Strains of an Infectious Disease. Bull Math Biol 2025; 87:19. [PMID: 39751961 DOI: 10.1007/s11538-024-01378-x] [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: 12/22/2023] [Accepted: 10/26/2024] [Indexed: 01/04/2025]
Abstract
We investigate the impact of differential vaccine effectiveness, waning immunity, and natural cross-immunity on the capacity for vaccine-induced strain replacement in two-strain models of infectious disease spread. We focus specifically on the case where the first strain is more transmissible but the second strain is more immune-resistant. We consider two cases on vaccine-induced immunity: (1) a monovalent model where the second strain has immune escape with respect to vaccination; and (2) a bivalent model where the vaccine remains equally effective against both strains. Our analysis reaffirms the capacity for vaccine-induced strain replacement under a variety of circumstances; surprisingly, however, we find that which strain is preferred depends sensitively on the degree of differential vaccine effectiveness. In general, the monovalent model favors the more immune-resistant strain at high vaccination levels while the bivalent model favors the more transmissible strain at high vaccination levels. To further investigate this phenomenon, we parametrize the bifurcation space between the monovalent and bivalent model.
Collapse
Affiliation(s)
- Matthew D Johnston
- Department of Mathematics and Computer Science, Lawrence Technological University, 21000 W. 10 Mile Rd., Southfield, MI, 48075, USA.
| | - Bruce Pell
- Department of Mathematics and Computer Science, Lawrence Technological University, 21000 W. 10 Mile Rd., Southfield, MI, 48075, USA
| | - Jared Pemberton
- Department of Mathematics and Computer Science, Lawrence Technological University, 21000 W. 10 Mile Rd., Southfield, MI, 48075, USA
| | - David A Rubel
- Department of Mathematics and Computer Science, Lawrence Technological University, 21000 W. 10 Mile Rd., Southfield, MI, 48075, USA
| |
Collapse
|
28
|
Stocks D, Thomas A, Finn A, Danon L, Brooks-Pollock E. Mechanistic models of humoral kinetics following COVID-19 vaccination. J R Soc Interface 2025; 22:20240445. [PMID: 39876790 PMCID: PMC11775660 DOI: 10.1098/rsif.2024.0445] [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: 07/01/2024] [Revised: 10/07/2024] [Accepted: 11/01/2024] [Indexed: 01/31/2025] Open
Abstract
COVID-19 vaccine programmes must account for variable immune responses and waning protection. Existing descriptions of antibody responses to COVID-19 vaccination convey limited information about the mechanisms of antibody production and maintenance. We describe antibody dynamics after COVID-19 vaccination with two biologically motivated mathematical models. We fit the models using Markov chain Monte Carlo to seroprevalence data from 14 602 uninfected individuals in England between May 2020 and September 2022. We analyse the effect of age, vaccine type, number of doses and the interval between doses on antibody production and longevity. We find evidence that individuals over 35 years old twice vaccinated with ChAdOx1-S generate a persistent antibody response suggestive of long-lived plasma cell induction. We also find that plasmablast productive capacity is greater in: younger people than older people (≤4.5-fold change in point estimates); people vaccinated with two doses than one dose (≤12-fold change); and people vaccinated with BNT162b2 than ChAdOx1-S (≤440-fold change). We find the half-life of an antibody to be 23-106 days. Routinely collected seroprevalence data are invaluable for characterizing within-host mechanisms of antibody production and persistence. Extended sampling and linking seroprevalence data to outcomes would enable conclusions about how humoral kinetics protect against disease.
Collapse
Affiliation(s)
- Daniel Stocks
- School of Engineering Mathematics and Technology, University of Bristol, Tankard’s Close, Bristol, BS8 1TW, UK
| | - Amy Thomas
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Adam Finn
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Leon Danon
- School of Engineering Mathematics and Technology, University of Bristol, Tankard’s Close, Bristol, BS8 1TW, UK
| | - Ellen Brooks-Pollock
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield Grove, Bristol, BS8 2BN, UK
| |
Collapse
|
29
|
Deng Y, Wang CS, Cheng GD, Whitson JA, Dow BJ, Lee AY. The role of interdependent self-construal in mitigating the effect of conspiratorial beliefs on vaccine acceptance. BRITISH JOURNAL OF SOCIAL PSYCHOLOGY 2025; 64:e12836. [PMID: 39651836 PMCID: PMC11627010 DOI: 10.1111/bjso.12836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024]
Abstract
Infectious diseases pose significant challenges to public health, leading to illness and even death. Vaccinations are vital for protecting society, yet beliefs in conspiracy theories related to infectious diseases increase vaccine hesitancy. This paper delves into vaccination decisions in the context of COVID-19, which continues to strain the health care system. While past research focuses on countering conspiratorial beliefs with cognitive persuasion interventions, we propose a social intervention as an alternative. Our novel intervention seeks to mitigate the effects of conspiratorial beliefs by fostering individuals' interdependent self-construal - viewing oneself in the context of social relationships. Interdependent self-construal was operationalized in multiple ways (measured in Studies 1, 2 and 3; manipulated to test causality in Studies 4 and 5). Conspiratorial beliefs were also manipulated in Study 5. The results show that the association between conspiratorial beliefs and vaccine hesitancy is weakened among individuals whose interdependent self-construal is more accessible. Moreover, this effect was mediated by prosocial motivation. We discuss the implications of our findings for developing and communicating health policies and propose potential contexts where this intervention may be relevant, thereby providing valuable insights into enhancing societal well-being in the face of conspiratorial beliefs.
Collapse
Affiliation(s)
- Yingli Deng
- Durham University Business SchoolDurham UniversityDurhamUK
| | - Cynthia S. Wang
- Kellogg School of ManagementNorthwestern UniversityEvanstonIllinoisUSA
| | | | - Jennifer A. Whitson
- Anderson School of ManagementUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Benjamin J. Dow
- Cox School of BusinessSouthern Methodist UniversityDallasTexasUSA
| | - Angela Y. Lee
- Kellogg School of ManagementNorthwestern UniversityEvanstonIllinoisUSA
| |
Collapse
|
30
|
Zhou G, Dael N, Verweij S, Balafas S, Mubarik S, Oude Rengerink K, Pasmooij AMG, van Baarle D, Mol PGM, de Bock GH, Hak E. Effectiveness of COVID-19 vaccines against SARS-CoV-2 infection and severe outcomes in adults: a systematic review and meta-analysis of European studies published up to 22 January 2024. Eur Respir Rev 2025; 34:240222. [PMID: 39971395 PMCID: PMC11836669 DOI: 10.1183/16000617.0222-2024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/11/2024] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Up-to-date evidence from European studies on long-term vaccine effectiveness (VE) of COVID-19 vaccines is lacking. This review aimed to evaluate effectiveness and durability of primary vaccine series and boosters in preventing infection and severe outcomes in the European population. METHODS We conducted systematic searches of PubMed and Embase up to 22 January 2024. We included observational studies that evaluated VE against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or severe disease (hospitalisation, intensive care unit admission or death) for primary series and boosters in Europe. We applied a random-effects meta-analysis model. RESULTS We included 33 studies and over 56 million participants. The overall VE of the complete primary series against infection with any SARS-CoV-2 variant was 70.7%. VE was lower for Omicron, at 26.1%, than for pre-Omicron strains, at 77.0%. Over time, VE against infection by any variant decreased from 68.9% to 38.9% after 6 months. Boosters restored VE to 76.4% and maintained at 58.4% after 3 months. The overall VE of a complete primary series for severe outcomes due to any variant was 87.4%, with 93.3% for pre-Omicron and 62.8% for Omicron strains. Protection against severe outcomes declined less than for infection. 6 months after the primary series, the vaccine still provided over 50% protection against severe outcomes caused by Omicron. Boosters restored VE to 87.9% and maintained at 78.5% after 3 months. CONCLUSION VE against SARS-CoV-2 infection declines markedly with time and Omicron variants. Protection against severe outcomes was more durable and resistant to viral mutation. Boosters restored protection, emphasising the need for timely booster vaccination for vulnerable populations.
Collapse
Affiliation(s)
- Guiling Zhou
- Unit of Pharmaco-Therapy, -Epidemiology and -Economics (PTEE), Department of Pharmacy, University of Groningen, Groningen, The Netherlands
- These authors contributed equally to this work
| | - Nina Dael
- Unit of Pharmaco-Therapy, -Epidemiology and -Economics (PTEE), Department of Pharmacy, University of Groningen, Groningen, The Netherlands
- These authors contributed equally to this work
| | - Stefan Verweij
- Unit of Pharmaco-Therapy, -Epidemiology and -Economics (PTEE), Department of Pharmacy, University of Groningen, Groningen, The Netherlands
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands
| | - Spyros Balafas
- Unit of Pharmaco-Therapy, -Epidemiology and -Economics (PTEE), Department of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Sumaira Mubarik
- Unit of Pharmaco-Therapy, -Epidemiology and -Economics (PTEE), Department of Pharmacy, University of Groningen, Groningen, The Netherlands
| | | | - Anna Maria Gerdina Pasmooij
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Debbie van Baarle
- Virology and Immunology Research Group, Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter G M Mol
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Geertruida H de Bock
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eelko Hak
- Unit of Pharmaco-Therapy, -Epidemiology and -Economics (PTEE), Department of Pharmacy, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
31
|
Alsulaiman JW, Alzoubi A, Alrawashdeh A, Al-Dekah AM, Abubaker S, Amayreh W, Sweileh WM, Alzoubi HM, Kheirallah KA. Mapping trends and hotspots of research on COVID-19 vaccine effectiveness: A comprehensive bibliometric analysis of global research. J Infect Public Health 2025; 18:102597. [PMID: 39603060 DOI: 10.1016/j.jiph.2024.102597] [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: 08/15/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND With an ever growing and expanding body of literature on the newly developed vaccines against the COVID-19, there is an urgent need for a comprehensive analysis of the current state of research on vaccine effectiveness (VE). This study conducted a comprehensive bibliometric analysis to critically examine the productivity and impact of retrieved publications on COVID-19 VE and to predict the future directions of research in the field. METHODS The global literature on COVID-19 VE from 2021 to 2024 was extracted from the VIEW-hub website. Using bibliometric analysis tools, specifically Microsoft Excel, the R package "bibliometrix, biblioshiny" and VOSviewer, we analyzed publications for trends in productivity, citations, and global collaboration. Key metrics assessed include publication and citation trends, influential authors, collaborative networks, and thematic evolution, offering a comprehensive view of the research landscape on COVID-19 VE. RESULTS A total of 490 publications were authored by 5031 authors from 934 institutions and 78 countries and published in 119 journals. Most retrieved publications were original articles (99.6 %). The United States was the most productive country with 205 publications (41.8 %). Global research collaborations were mainly within developed countries. Analysis of the thematic evolution of the field illustrated changing research focus over three distinct time clusters. Throughout 2021, studies were focused on outlining infection prevention and control measures, as well as examining the efficacy of novel mRNA vaccines. In 2022, the linchpin of research was shifted towards dissecting the epidemiological correlates of the pandemic in light of the widespread use of vaccines. The final cluster showed special emphasis on the new variants of COVID-19 and the long-term outcomes of vaccines. CONCLUSION Our study identified geopolitical disparities and weak engagement from developing countries in the ongoing efforts regarding COVID-19 VE. This study can inform researchers, policymakers, and funding agencies as they assess ongoing research and future directions in COVID-19 VE.
Collapse
Affiliation(s)
- Jomana W Alsulaiman
- Department of Pediatrics, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Abdallah Alzoubi
- Department of Pharmacology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Ahmad Alrawashdeh
- Department of Allied Medical Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | | | - Sara Abubaker
- Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Wajdi Amayreh
- Department of Pediatrics, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Waleed M Sweileh
- Department of Physiology and Pharmacology/Toxicology, College of Medicine and Sciences, An-Najah National University, Nablus, Palestine
| | - Hamed M Alzoubi
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Khalid A Kheirallah
- Department of Public Health and Family Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan.
| |
Collapse
|
32
|
Hassan MZ, Basher AK, Rahman MZ, Bhuiyan TR, Chowdhury F, Hossain MK, Rahman A, Islam MN, M Duca L, Kaydos-Daniels SC, A Dahl B, Qadri F, Ortiz N. Study protocol for COVID-19 breakthrough infections and vaccine-induced immune response among a cohort of healthcare workers, Bangladesh. PLoS One 2024; 19:e0316121. [PMID: 39739828 DOI: 10.1371/journal.pone.0316121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/04/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND To optimize vaccination strategies, it is useful to detect breakthrough infections and assess vaccine effectiveness in programmatic use. Monitoring emerging SARS-CoV-2 variants and vaccine effectiveness against them is also essential to determine the most effective vaccine options. This study aims to monitor SARS-CoV-2 breakthrough infections, the emergence of new SARS-CoV-2 variants, and host immune response during the peri-infection period of COVID-19. The study will also assess the uptake of the COVID-19 vaccine booster doses, and associated barriers or motivations among healthcare workers (HCWs). METHODS Leveraging an existing HCW cohort in Bangladesh, HCWs will be enrolled from purposively selected health facilities from four different administrative divisions across Bangladesh. We captured cohort data on HCW's demographic information, clinical information, COVID-19 illness, and exposure, and vaccination histories for COVID-19. However, no biological specimens were collected for testing during the first phase of the cohort. In the current study, we plan to follow enrolled HCWs biweekly for suspected COVID-19 illness and capture relevant data including illness outcomes. Respiratory swab samples from symptomatic and a subset of asymptomatic HCWs will be tested for SARS-CoV-2 by rRT-PCR and positive samples will undergo Sanger sequencing to identify the SARS-CoV-2 variants of concern (VOCs). We will also perform Whole Genome Sequencing on a subset of SARS-CoV-2 positive samples with low CT values (Ct ≤ 30) to identify emerging SARS-CoV-2 variants. To examine the antibody response, we will collect blood samples from the participants at 12-week intervals for one year. We will use the EUROIMMUN kit and will also perform in-house ELISA to assess host immune factors with Luminex platform. DISCUSSION This proposed study will generate useful data on COVID-19 breakthrough infection and the durability of anti-SARS-CoV-2 antibodies among HCWs following vaccination. The findings on booster vaccination intention and uptake will inform government COVID-19 vaccination strategies. Information on circulating and emerging strains of SARS-CoV-2 and vaccine performance against those strains will help understand population-level risks of COVID-19 infection. The study will generate data on facilitators and barriers to COVID-19 booster uptake among HCWs which can inform health communication messaging to improve booster acceptance in this population.
Collapse
Affiliation(s)
- Md Zakiul Hassan
- Infectious Disease Division, Programme for Emerging Infections, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Ahamed Khairul Basher
- Infectious Disease Division, Programme for Emerging Infections, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mohammed Ziaur Rahman
- Infectious Disease Division, Programme for Emerging Infections, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Taufiqur Rahman Bhuiyan
- Infectious Disease Division, Programme for Emerging Infections, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Fahmida Chowdhury
- Infectious Disease Division, Programme for Emerging Infections, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md Kamal Hossain
- Infectious Disease Division, Programme for Emerging Infections, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Aninda Rahman
- Communicable Disease Control, the Director General of Health Services, Ministry of Health and Family Welfare Government of Bangladesh, Dhaka, Bangladesh
| | - Md Nazmul Islam
- Communicable Disease Control, the Director General of Health Services, Ministry of Health and Family Welfare Government of Bangladesh, Dhaka, Bangladesh
| | - Lindsey M Duca
- US Centers for Disease Control and Prevention, Atlanta, GA, United States of America
| | | | - Benjamin A Dahl
- US Centers for Disease Control and Prevention, Atlanta, GA, United States of America
| | - Firdausi Qadri
- Infectious Disease Division, Programme for Emerging Infections, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Nancy Ortiz
- US Centers for Disease Control and Prevention, Atlanta, GA, United States of America
| |
Collapse
|
33
|
Petrovsky N. Clinical development of SpikoGen®, an Advax-CpG55.2 adjuvanted recombinant spike protein vaccine. Hum Vaccin Immunother 2024; 20:2363016. [PMID: 38839044 PMCID: PMC11155708 DOI: 10.1080/21645515.2024.2363016] [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: 02/01/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
Recombinant protein vaccines represent a well-established, reliable and safe approach for pandemic vaccination. SpikoGen® is a recombinant spike protein trimer manufactured in insect cells and formulated with Advax-CpG55.2 adjuvant. In murine, hamster, ferret and non-human primate studies, SpikoGen® consistently provided protection against a range of SARS-CoV-2 variants. A pivotal Phase 3 placebo-controlled efficacy trial involving 16,876 participants confirmed the ability of SpikoGen® to prevent infection and severe disease caused by the virulent Delta strain. SpikoGen® subsequently received a marketing authorization from the Iranian FDA in early October 2021 for prevention of COVID-19 in adults. Following a successful pediatric study, its approval was extended to children 5 years and older. Eight million doses of SpikoGen® have been delivered, and a next-generation booster version is currently in development. This highlights the benefits of adjuvanted protein-based approaches which should not overlook when vaccine platforms are being selected for future pandemics.
Collapse
Affiliation(s)
- Nikolai Petrovsky
- Research Department, Australian Respiratory and Sleep Medicine Institute Ltd, Adelaide, Australia
- Research Department, Vaxine Pty Ltd, Warradale, Australia
| |
Collapse
|
34
|
Yang X, Shi F, Zhang J, Gao H, Chen S, Olatosi B, Weissman S, Li X. Vaccination status and disease severity of COVID-19 in different phases of the pandemic. Hum Vaccin Immunother 2024; 20:2353491. [PMID: 38832632 PMCID: PMC11152109 DOI: 10.1080/21645515.2024.2353491] [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: 01/24/2024] [Accepted: 05/07/2024] [Indexed: 06/05/2024] Open
Abstract
This study aimed to explore the clinical profile and the impact of vaccination status on various health outcomes among COVID-19 patients diagnosed in different phases of the pandemic, during which several variants of concern (VOCs) circulated in South Carolina (SC). The current study included 861,526 adult COVID-19 patients diagnosed between January 2021 and April 2022. We extracted their information about demographic characteristics, vaccination, and clinical outcomes from a statewide electronic health record database. Multiple logistic regression models were used to compare clinical outcomes by vaccination status in different pandemic phases, accounting for key covariates (e.g. historical comorbidities). A reduction in mortality was observed among COVID-19 patients during the whole study period, although there were fluctuations during the Delta and Omicron dominant periods. Compared to non-vaccinated patients, full-vaccinated COVID-19 patients had lower mortality in all dominant variants, including Pre-alpha (adjusted odds ratio [aOR]: 0.33; 95%CI: 0.15-0.72), Alpha (aOR: 0.58; 95%CI: 0.42-0.82), Delta (aOR: 0.28; 95%CI: 0.25-0.31), and Omicron (aOR: 0.29; 95%CI: 0.26-0.33) phases. Regarding hospitalization, full-vaccinated parties showed lower risk of hospitalization than non-vaccinated patients in Delta (aOR: 0.44; 95%CI: 0.41-0.47) and Omicron (aOR: 0.53; 95%CI: 0.50-0.57) dominant periods. The findings demonstrated the protection effect of the COVID-19 vaccines against all VOCs, although some of the full-vaccinated population still have symptoms to varying degrees from COVID-19 disease at different phases of the pandemic.
Collapse
Affiliation(s)
- Xueying Yang
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Department of Health Promotion, Education and Behavior, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Fanghui Shi
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Department of Health Promotion, Education and Behavior, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Jiajia Zhang
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Haoyuan Gao
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Shujie Chen
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Bankole Olatosi
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Department of Health Services Policy and Management, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Sharon Weissman
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Department of Internal Medicine, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Xiaoming Li
- South Carolina SmartState Center for Healthcare Quality, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Department of Health Promotion, Education and Behavior, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
35
|
Stowe J, Lopez-Bernal J, Andrews N. The risk of acute disseminated encephalomyelitis (ADEM) following covid-19 vaccination in England: A self-controlled case-series analysis. Hum Vaccin Immunother 2024; 20:2311969. [PMID: 38299507 PMCID: PMC10841003 DOI: 10.1080/21645515.2024.2311969] [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: 07/19/2023] [Accepted: 01/26/2024] [Indexed: 02/02/2024] Open
Abstract
Acute disseminated encephalomyelitis (ADEM) has been identified as an Adverse Event of Special Interest in the COVID-19 vaccine programme due to its long-standing temporal association with a wide range of other vaccines. Case reports of ADEM shortly following COVID-19 vaccination have now been documented. There were 217 ADEM admissions in 215 individuals in the period 8th December 2020 to 31st March 2023. An increased risk of ADEM following the first dose of ChAdOx1 vaccine was observed (relative incidence (RI) = 3.13, 95% Confidence Interval (CI) [1.56-6.25]) with a vaccine attributable risk of 0.39 per million doses. When doses 1 and 2 were combined this increased risk remained just significant (1.96 [95%CI 1.01-3.82]). No significant increased risk was observed with any other vaccine or dose. This small, elevated risk after the first dose of ChAdOx1-S vaccine demonstrates how large national electronic datasets can be used to identify very rare risks and provides reassurance that any risk of ADEM following the ChAdOx1-S COVID-19 vaccination is extremely small. Given the rarity of this risk, further studies in settings with access to data on large populations should be carried out to verify these findings.
Collapse
Affiliation(s)
- Julia Stowe
- Immunisation Division, UK Health Security Agency, London, UK
| | | | - Nick Andrews
- Immunisation Division, UK Health Security Agency, London, UK
| |
Collapse
|
36
|
Rustagi V, Gupta SRR, Talwar C, Singh A, Xiao ZZ, Jamwal R, Bala K, Bhaskar AK, Nagar S, Singh IK. SARS-CoV-2 pathophysiology and post-vaccination severity: a systematic review. Immunol Res 2024; 73:17. [PMID: 39692912 DOI: 10.1007/s12026-024-09553-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/02/2024] [Indexed: 12/19/2024]
Abstract
Currently, COVID-19 is still striking after 4 years of prevalence, with millions of cases and thousands of fatalities being recorded every month. The virus can impact other major organ systems, including the gastrointestinal tract (GIT), cardiovascular, central nervous system, renal, and hepatobiliary systems. The resulting organ dysfunction from SARS-CoV-2 may be attributed to one or a combination of mechanisms, such as direct viral toxicity, disruptions in the renin-angiotensin-aldosterone system (RAAS), thrombosis, immune dysregulation, and ischemic injury due to vasculitis. SARS-CoV-2 vaccines effectively reduce the severity of the disease, hospitalizations, and mortality. As of October 2024, 13.58 billion vaccine doses have been administered, with an average of 6959 daily doses. Also, the boosters are given after the primary immunization in a homologous and heterologous manner. The vaccines imposed severe potential health side effects such as clotting or obstruction of blood vessels termed arterial or venous thrombosis, autoimmune damage of nerve cells (Guillain-Barré syndrome; GBS), intense activation of coagulation system (vaccine-induced thrombotic thrombocytopenia), acute ischemic stroke (AIS) and cerebral venous sinus thrombosis (CVST), myocarditis, pericarditis, and glomerular disease. Overall, it is essential to highlight that the significant benefits of COVID-19 vaccination far outweigh the low risk of conditions. mRNA-based vaccine technology has emerged as a rapidly deployable vaccine candidate and a viable alternative to existing vaccines. It has a very low probability of adverse health effects, confirmed by data represented by Preferred Reporting Items for Systematic Reviews and Meta-Analyses, Vaccine Adverse Event Reporting System (VAERS), Yellow card approved under CDC, WHO.
Collapse
Affiliation(s)
- Vanshika Rustagi
- Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, 110019, India
| | - Shradheya R R Gupta
- Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, 110019, India
| | - Chandni Talwar
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Archana Singh
- Department of Plant Molecular Biology, University of Delhi (South Campus), New Delhi, 110021, India
| | - Zhen-Zhu Xiao
- Department of Biological Sciences, The George Washington University, Washington, D.C, 20052, USA
| | - Rahul Jamwal
- Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, 110019, India
| | - Kiran Bala
- Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, 110019, India
| | - Akash Kumar Bhaskar
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, 110025, India
| | - Shekhar Nagar
- Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, 110019, India.
| | - Indrakant K Singh
- Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, 110019, India.
- Delhi School of Public Health, Institute of Eminence, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
37
|
Galmiche S, Coustaury C, Charniga K, Grant R, Cauchemez S, Fontanet A. Patterns and drivers of excess mortality during the COVID-19 pandemic in 13 Western European countries. BMC GLOBAL AND PUBLIC HEALTH 2024; 2:78. [PMID: 39681939 DOI: 10.1186/s44263-024-00103-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/09/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND Important differences in excess mortality between European countries during the COVID-19 pandemic have been reported. Understanding the drivers of these differences is essential to pandemic preparedness. METHODS We examined patterns in age- and sex-standardized cumulative excess mortality in 13 Western European countries during the first 30 months of the COVID-19 pandemic and the correlation of country-level characteristics of interest with excess mortality. RESULTS In a timeline analysis, we identified notable differences in seeding events, particularly in early 2020 and when the Alpha variant emerged, likely contributing to notable differences in excess mortality between countries (lowest in Denmark during that period). These differences were more limited from July 2021 onwards. Lower excess mortality was associated with implementing stringent non-pharmaceutical interventions (NPIs) when hospital admissions were still low in 2020 (correlation coefficient rho = 0.65, p = 0.03) and rapid rollout of vaccines in the elderly in early 2021 (rho = - 0.76, p = 0.002). Countries which implemented NPIs while hospital admissions were low tended to experience lower gross domestic product (GDP) losses in 2020 (rho = - 0.55, p = 0.08). Structural factors, such as high trust in the national government (rho = - 0.77, p = 0.002) and low ratio of population at risk of poverty (rho = 0.55, p = 0.05), were also associated with lower excess mortality. CONCLUSIONS These results suggest the benefit of early implementation of NPIs and swift rollout of vaccines to the most vulnerable. Further analyses are required at a more granular level to better understand how these factors impacted excess mortality and help guide pandemic preparedness plans.
Collapse
Affiliation(s)
- Simon Galmiche
- Emerging Diseases Epidemiology Unit, Institut Pasteur, Université Paris Cité, 25 Rue du Docteur Roux, 75015, Paris, France
| | - Camille Coustaury
- Emerging Diseases Epidemiology Unit, Institut Pasteur, Université Paris Cité, 25 Rue du Docteur Roux, 75015, Paris, France
| | - Kelly Charniga
- Mathematical Modelling of Infectious Diseases Unit, UMR2000, Institut Pasteur, Université Paris Cité, CNRS, Paris, France
| | - Rebecca Grant
- Infection Control Program and WHO Collaborating Centre, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Simon Cauchemez
- Mathematical Modelling of Infectious Diseases Unit, UMR2000, Institut Pasteur, Université Paris Cité, CNRS, Paris, France
| | - Arnaud Fontanet
- Emerging Diseases Epidemiology Unit, Institut Pasteur, Université Paris Cité, 25 Rue du Docteur Roux, 75015, Paris, France.
- Conservatoire National Des Arts Et Métiers, Unité PACRI, Paris, France.
| |
Collapse
|
38
|
Kirsebom FCM, Andrews N, Mensah AA, Stowe J, Ladhani S, Ramsay M, Lopez Bernal J, Campbell HJ. Vaccine effectiveness against mild and severe covid-19 in pregnant individuals and their infants in England: test negative case-control study. BMJ MEDICINE 2024; 3:e000696. [PMID: 39902238 PMCID: PMC11789471 DOI: 10.1136/bmjmed-2023-000696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/14/2024] [Indexed: 02/05/2025]
Abstract
Objective To estimate real world vaccine effectiveness against symptomatic disease and hospital admission with the delta and omicron variants of the SARS-CoV-2 virus in pregnant individuals, and to estimate the protection conferred by previous infection and maternal vaccination in their infants. Design Test negative case-control study. Setting Community and hospital testing for covid-19, in England, 26 April 2021 to 9 January 2022 (delta variant period) and 29 November 2021 to 31 March 2022 (omicron variant period). Testing data were linked to Hospital Episode Statistics and Maternal Services Data Set (for data on pregnant individuals and infants), National Immunisation Management System (for covid-19 vaccinations), and Secondary Uses Service (for hospital admissions). Participants 35 206 negative and 16 693 positive eligible test results in the delta variant period from pregnant individuals with symptoms of infection, aged 16-55 years, whose pregnancy ended in 2021, and 5974 negative and 4715 positive eligible test results in the omicron variant period. For infants born in 2021, 23 053 negative and 2924 positive eligible test results in the delta variant period and 13 908 negative and 5669 positive test results from infants in the omicron period. Main outcome measures Vaccine effectiveness against symptomatic disease and hospital admission with the delta and omicron variants of the SARS-CoV-2 virus in pregnant women. Also, effectiveness of maternal vaccination and the protection conferred by previous infection in mothers in preventing symptomatic disease and hospital admission in their infants in the first six months of life. Symptomatic SARS-CoV-2 infection was confirmed by a positive polymerase chain reaction test result. Results Vaccine effectiveness against symptomatic disease (delta and omicron infection) and against hospital admission (delta infection only) in pregnant individuals was high, as seen in the general population. A booster dose of vaccine gave sustained protection, with no evidence of waning up to 15 weeks after vaccination. Vaccine effectiveness against symptomatic disease peaked at 98.4% (95% confidence interval (CI) 88.4% to 99.8%) and 80.1% (73.8% to 84.9%) against the delta and omicron variants, respectively, after the booster dose of vaccine. Vaccine effectiveness after a two dose primary schedule against hospital admission with delta infection peaked at 92.7% (95% CI 79.9% to 97.4%) in pregnant individuals. Maternal vaccination during and after pregnancy also provided sustained protection from symptomatic disease and hospital admission after delta and omicron infection in infants aged up to six months, with the highest protection seen when maternal vaccination occurred during later pregnancy. The effectiveness of two maternal doses when the last dose was given in the third trimester was 86.5% (95% CI 81.9% to 90.0%) and 56.6% (46.7% to 64.6%) against symptomatic disease with delta and omicron infection, respectively, in infants, and effectiveness against hospital admission was 94.7% (78.2% to 98.7%) and 78.7% (58.2% to 89.1%), respectively. Previous infection with wild-type, alpha, and delta variants of the SARS-CoV-2 virus in pregnant individuals was more protective against mild and severe delta infection than omicron infection in their infants. Conclusions The results of this study indicated that maternal vaccination prevented mild and severe disease in pregnant individuals and their infants for up to six months after birth. The findings support the promotion of both primary and booster vaccination for pregnant individuals to protect themselves and their infants.
Collapse
Affiliation(s)
- Freja C M Kirsebom
- Immunisation and
Vaccine Preventable Diseases, UK Health Security
Agency, London,
UK
| | - Nick Andrews
- Immunisation and
Vaccine Preventable Diseases, UK Health Security
Agency, London,
UK
- London School of
Hygiene and Tropical Medicine, National Institute for Health
and Care Research (NIHR) Health Protection Research Unit in Vaccines and
Immunisation, London,
UK
| | - Anna A Mensah
- Immunisation and
Vaccine Preventable Diseases, UK Health Security
Agency, London,
UK
| | - Julia Stowe
- Immunisation and
Vaccine Preventable Diseases, UK Health Security
Agency, London,
UK
| | - Shamez Ladhani
- Immunisation and
Vaccine Preventable Diseases, UK Health Security
Agency, London,
UK
- St George’s University of
London, London,
UK
| | - Mary Ramsay
- Immunisation and
Vaccine Preventable Diseases, UK Health Security
Agency, London,
UK
- London School of
Hygiene and Tropical Medicine, National Institute for Health
and Care Research (NIHR) Health Protection Research Unit in Vaccines and
Immunisation, London,
UK
| | - Jamie Lopez Bernal
- Immunisation and
Vaccine Preventable Diseases, UK Health Security
Agency, London,
UK
- London School of
Hygiene and Tropical Medicine, National Institute for Health
and Care Research (NIHR) Health Protection Research Unit in Vaccines and
Immunisation, London,
UK
- Imperial College
London, National Institute for Health and Care Research
(NIHR) Health Protection Research Unit in Respiratory Infections,
London, UK
| | - Helen Julia Campbell
- Immunisation and
Vaccine Preventable Diseases, UK Health Security
Agency, London,
UK
| |
Collapse
|
39
|
Tsang TK, Sullivan SG, Huang X, Wang C, Wang Y, Nealon J, Yang B, Ainslie KEC, Cowling BJ. Prior infections and effectiveness of SARS-CoV-2 vaccine in test-negative studies: a systematic review and meta-analysis. Am J Epidemiol 2024; 193:1868-1881. [PMID: 38904437 PMCID: PMC11637527 DOI: 10.1093/aje/kwae142] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/17/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024] Open
Abstract
Prior infection with SARS-CoV-2 can provide protection against infection and severe COVID-19. We aimed to determine the impact of preexisting immunity on vaccine effectiveness (VE) estimates. We systematically reviewed and meta-analyzed 66 test-negative design studies that examined VE against infection or severe disease (hospitalization, intensive care unit admission, or death) for primary vaccination series. Pooled VE among studies that included people with prior COVID-19 infection was lower against infection (77%; 95% CI, 72-81) and severe disease (86%; 95% CI, 83-89) compared with studies that excluded people with prior COVID-19 infection (pooled VE against infection: 87% [95% CI, 85-89]; pooled VE against severe disease: 93% [95% CI, 91-95]). There was a negative correlation between VE estimates against infection and severe disease, and the cumulative incidence of cases before the start of the study or incidence rates during the study period. We found clear empirical evidence that higher levels of preexisting immunity were associated with lower VE estimates. Prior infections should be treated as both a confounder and effect modificatory when the policies target the whole population or are stratified by infection history, respectively.
Collapse
Affiliation(s)
- Tim K Tsang
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| | - Sheena G Sullivan
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, and Doherty Department, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Xiaotong Huang
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Can Wang
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yifan Wang
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Joshua Nealon
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Bingyi Yang
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kylie E C Ainslie
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Centre for Infectious Disease Control, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - Benjamin J Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| |
Collapse
|
40
|
Meakin S, Nsio J, Camacho A, Kitenge R, Coulborn RM, Gignoux E, Johnson J, Sterk E, Musenga EM, Mustafa SHB, Finger F, Ahuka-Mundeke S. Effectiveness of rVSV-ZEBOV vaccination during the 2018-20 Ebola virus disease epidemic in the Democratic Republic of the Congo: a retrospective test-negative study. THE LANCET. INFECTIOUS DISEASES 2024; 24:1357-1365. [PMID: 39178866 DOI: 10.1016/s1473-3099(24)00419-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/08/2024] [Accepted: 06/20/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND The recombinant vesicular stomatitis virus-Zaire Ebola virus (rVSV-ZEBOV) vaccine is the only WHO prequalified vaccine recommended for use to respond to outbreaks of Ebola virus (species Zaire ebolavirus) by WHO's Strategic Advisory Group of Experts on Immunization. Despite the vaccine's widespread use during several outbreaks, no real-world effectiveness estimates are currently available in the literature. METHODS We conducted a retrospective test-negative analysis to estimate effectiveness of rVSV-ZEBOV vaccination against Ebola virus disease during the 2018-20 epidemic in the Democratic Republic of the Congo, using data on suspected Ebola virus disease cases collected from Ebola treatment centres. Those eligible for inclusion had an available Ebola virus RT-PCR result, available key data, were eligible for vaccination during the outbreak, and had symptom onset aligning with the period in which a ring-vaccination protocol was in use. After imputing missing data, each individual confirmed by RT-PCR to be Ebola virus disease-positive (defined as a case) was matched to one individual negative for Ebola virus disease (control) by sex, age, health zone, and month of symptom onset. Effectiveness was estimated from the odds ratio of being vaccinated (≥10 days before symptom onset) versus being unvaccinated among cases and controls, after adjusting for the matching factors. The imputation, matching and effectiveness estimation, was repeated 500 times. FINDINGS 1273 (4·8%) of 26 438 eligible individuals were positive for Ebola virus disease (cases) and 25 165 (95·2%) were negative (controls). 40 (3·1%) cases and 1271 (5·1%) controls were reported as being vaccinated at least 10 days before symptom onset. After selecting individuals who reported exposure to an individual with Ebola virus disease within the 21 days before symptom onset and matching, the analysis datasets comprised a median of 309 cases and 309 controls. 10 days or more after vaccination, the effectiveness of rVSV-ZEBOV against Ebola virus disease was estimated to be 84% (95% credible interval 70-92). INTERPRETATION This analysis is the first to provide estimates of the real-world effectiveness of the rVSV-ZEBOV vaccine against Ebola virus disease, amid the widespread use of the vaccine during a large Ebola virus disease outbreak. Our findings confirm that rVSV-ZEBOV is highly protective against Ebola virus disease and support its use during outbreaks, even in challenging contexts such as in the eastern Democratic Republic of the Congo. FUNDING Médecins Sans Frontières. TRANSLATION For the French translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
| | - Justus Nsio
- General Direction of Disease Control, Ministry of Public Health, Hygiene, and Prevention, Kinshasa, Democratic Republic of the Congo
| | | | - Richard Kitenge
- National Program of Care and Follow-up of Survivors, Ministry of Public Health, Hygiene, and Prevention, Kinshasa, Democratic Republic of the Congo
| | | | | | | | | | - Elisabeth Mukamba Musenga
- Expanded Programme on Immunization, Ministry of Public Health, Hygiene, and Prevention, Kinshasa, Democratic Republic of the Congo
| | - Stephane Hans Bateyi Mustafa
- Expanded Programme on Immunization, Ministry of Public Health, Hygiene, and Prevention, Goma, Democratic Republic of the Congo; Department of Public Health, Faculty of Medicine, University of Goma, Goma, Democratic Republic of the Congo; Department of Epidemiology, Faculty of Health and Community Development, Université de Pays de Grand Lacs, Goma, Democratic Republic of the Congo
| | | | - Steve Ahuka-Mundeke
- Department of Virology, Institut National de la Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Department of Medical Biology, Cliniques Universitaires de Kinshasa, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| |
Collapse
|
41
|
Dings C, Selzer D, Bragazzi NL, Möhler E, Wenning M, Gehrke T, Richter U, Nonnenmacher A, Brinkmann F, Rothoeft T, Zemlin M, Lücke T, Lehr T. Effect of vaccinations and school restrictions on the spread of COVID-19 in different age groups in Germany. Infect Dis Model 2024; 9:1250-1264. [PMID: 39183948 PMCID: PMC11342094 DOI: 10.1016/j.idm.2024.07.004] [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: 04/01/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
With the emergence of SARS-CoV-2, various non-pharmaceutical interventions were adopted to control virus transmission, including school closures. Subsequently, the introduction of vaccines mitigated not only disease severity but also the spread of SARS-CoV-2. This study leveraged an adapted SIR model and non-linear mixed-effects modeling to quantify the impact of remote learning, school holidays, the emergence of Variants of Concern (VOCs), and the role of vaccinations in controlling SARS-CoV-2 spread across 16 German federal states with an age-stratified approach. Findings highlight a significant inverse correlation (Spearman's ρ = -0.92, p < 0.001) between vaccination rates and peak incidence rates across all age groups. Model-parameter estimation using the observed number of cases stratified by federal state and age allowed to assess the effects of school closure and holidays, considering adjustments for vaccinations and spread of VOCs over time. Here, modeling revealed significant (p < 0.001) differences in the virus's spread among pre-school children (0-4), children (5-11), adolescents (12-17), adults (18-59), and the elderly (60+). The transition to remote learning emerged as a critical measure in significantly reducing infection rates among children and adolescents (p < 0.001), whereas an increased infection risk was noted among the elderly during these periods, suggesting a shift in infection networks due to altered caregiving roles. Conversely, during school holiday periods, infection rates among adolescents mirrored those observed when schools were open. Simulation exercises based on the model provided evidence that COVID-19 vaccinations might serve a dual purpose: they protect the vaccinated individuals and contribute to the broader community's safety.
Collapse
Affiliation(s)
- Christiane Dings
- Department of Clinical Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| | - Dominik Selzer
- Department of Clinical Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| | | | - Eva Möhler
- Department of Child and Adolescent Psychiatry, Saarland University Hospital, 66421, Homburg, Germany
| | - Markus Wenning
- Medical Association, Westfalen-Lippe, 48151, Münster, Germany
| | - Thomas Gehrke
- Medical Association, Westfalen-Lippe, 48151, Münster, Germany
| | - Ulf Richter
- School of Education and Psychology, Siegen University, 57072, Siegen, Germany
| | | | - Folke Brinkmann
- University Children's Hospital, Ruhr University, 44791, Bochum, Germany
- University Children's Hospital, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Lübeck, Germany
| | - Tobias Rothoeft
- University Children's Hospital, Ruhr University, 44791, Bochum, Germany
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Saarland University Hospital, 66421, Homburg, Germany
| | - Thomas Lücke
- Medical Association, Westfalen-Lippe, 48151, Münster, Germany
- University Children's Hospital, Ruhr University, 44791, Bochum, Germany
| | - Thorsten Lehr
- Department of Clinical Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| |
Collapse
|
42
|
Nakatani E, Morioka H, Kikuchi T, Fukushima M. Behavioral and Health Outcomes of mRNA COVID-19 Vaccination: A Case-Control Study in Japanese Small and Medium-Sized Enterprises. Cureus 2024; 16:e75652. [PMID: 39803093 PMCID: PMC11725330 DOI: 10.7759/cureus.75652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Background Despite ongoing waves of Coronavirus disease 2019 (COVID-19) infections, including significant surges such as the 10th wave, understanding the impact of messenger RNA (mRNA) COVID-19 vaccination on infection risk and associated behavioral changes remains crucial. This study aims to urgently evaluate the effects of mRNA COVID-19 vaccination on COVID-19 infection rates and related behaviors among participants of the Yamato Project, which includes employees of Japanese small and medium-sized enterprises (SMEs). Methods A case-control study was conducted using data collected from a survey administered by the Japan Small and Medium Enterprise Management Council in December 2023. Participants included individuals who were part of the Yamato Project, not necessarily limited to SME employees. The survey gathered information on demographic characteristics, COVID-19 infection status, vaccination history, health status before January 2020, and various preventive behaviors. The primary outcome was the presence or absence of COVID-19 infection. Data were analyzed using univariate and multivariate logistic regression models to calculate odds ratios (ORs) and 95% confidence intervals (CIs) for the association between vaccination status and COVID-19 infection. Results A total of 913 participants were included in the final analysis. The adjusted ORs for COVID-19 infection among vaccinated individuals compared to unvaccinated individuals were 1.85 (95% CI: 1.33-2.57, p < 0.001). The odds of contracting COVID-19 increased with the number of vaccine doses: one to two doses (OR: 1.63, 95% CI: 1.08-2.46, p = 0.020), three to four doses (OR: 2.04, 95% CI: 1.35-3.08, p = 0.001), and five to seven doses (OR: 2.21, 95% CI: 1.07-4.56, p = 0.033). Behavioral analysis indicated that a reduced frequency of bathing and exercising was significantly associated with higher COVID-19 infection rates (p < 0.05). Conclusions The study observed a higher reported incidence of COVID-19 infection among vaccinated individuals during the pandemic period, which increased with the number of vaccine doses received. This paradoxical finding may be influenced by various factors, including immune response mechanisms, such as antibody-dependent enhancement (ADE) or original antigenic sin, behavioral changes, and exposure risk. Understanding these factors is crucial for urgently enhancing public health strategies and vaccination programs.
Collapse
Affiliation(s)
- Eiji Nakatani
- Research Support Center, Shizuoka General Hospital, Shizuoka, JPN
| | - Hisayo Morioka
- Health Data Science, Learning Health Society Institute, Nagoya, JPN
| | - Takayuki Kikuchi
- Health Data Science, Learning Health Society Institute, Nagoya, JPN
| | | |
Collapse
|
43
|
Kim J, Jung H. Statistical Analysis of Multiple Vaccine Effectiveness Against COVID-19 Variants: Integrating Immune Response Dynamics. Cureus 2024; 16:e76705. [PMID: 39898140 PMCID: PMC11782921 DOI: 10.7759/cureus.76705] [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: 12/31/2024] [Indexed: 02/04/2025] Open
Abstract
The study aims to evaluate the association between multiple COVID-19 vaccine doses and daily confirmed cases, incorporating immune response delays to better understand vaccination efficacy. We investigated the effectiveness of multiple COVID-19 vaccinations, with particular emphasis on viral variants, by integrating immune response dynamics into the statistical analysis to provide a more accurate assessment of vaccination outcomes. Detailed data on vaccination numbers and confirmed COVID-19 cases were obtained from the official Korean Ministry of the Interior and Safety website. Multiple linear regression was applied to evaluate the effectiveness of each vaccine dose against different COVID-19 variants. This approach offered a clearer understanding of how vaccination influenced the spread of COVID-19 over time. By accounting for delay periods associated with immune responses, we aimed to provide more accurate insights into the effectiveness of each vaccine dose. The results underscored the impact of multiple vaccinations when incorporating immune response timing and revealed notable differences in the responses of the Delta and Omicron variants to multiple doses. While significant effectiveness was observed against the Omicron variant, a similar positive effect was not evident for the Delta variant.
Collapse
Affiliation(s)
- Jiwoo Kim
- Medicine, John A. Burns School of Medicine, University of Hawai'i at Mānoa, Honolulu, USA
| | - Hyosoon Jung
- Preventive Medicine, Defense Health Agency, Area IV, Daegu, KOR
| |
Collapse
|
44
|
Su YCF, Zeller MA, Ou TP, Ma J, Pum L, Zhang R, Rath S, Heang V, Kol S, Lim R, Chea KL, Khun L, Heng L, Krang S, Raftery P, Kinzer MH, Ieng V, Kab V, Patel S, Sar B, Horm VS, Yann S, Auerswald H, Siegers JY, Troupin C, Boukli N, Vandelannoote K, Wong FY, Ng GGK, Chan M, Sorn S, Sengdoeurn Y, Heng S, Darapheak C, Savuth C, Khalakdina A, Ly S, Baril L, Spiegel A, Duong V, Ly S, Smith GJD, Karlsson EA. Spatiotemporal evolution and transmission dynamics of Alpha and Delta SARS-CoV-2 variants contributing to sequential outbreaks in Cambodia during 2021. COMMUNICATIONS MEDICINE 2024; 4:252. [PMID: 39604601 PMCID: PMC11603031 DOI: 10.1038/s43856-024-00685-7] [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: 11/22/2023] [Accepted: 11/20/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Tracking the emergence, introduction and spread of SARS-CoV-2 variants of concern are essential for informing public health strategies. In 2021, Cambodia faced two major epidemic waves of SARS-CoV-2 triggered by the successive rise of the Alpha and Delta variants. METHODS Phylodynamic analysis of 1,163 complete SARS-CoV-2 genomes from Cambodia, along with global sequences, were conducted between February and September 2021 to infer viral introductions, molecular epidemiology and population dynamics. The relationship between epidemic trends and control strategies were evaluated. Bayesian phylogeographic reconstruction was employed to estimate and contrast the spatiotemporal dynamics of the Alpha and Delta variants over time. RESULTS Here we reveal that the Alpha variant displays rapid lineage diversification, accompanied by the acquisition of a spike E484K mutation that coincides with the national implementation of mass COVID-19 vaccination. Despite nationwide control strategies and increased vaccination coverage, the Alpha variant was quickly displaced by Delta variants that exhibits a higher effective reproductive number. Phylogeographic inference indicates that the Alpha variant was introduced through south-central region of Cambodia, with strong diffusion rates from the capital of Phnom Penh to other provinces, while the Delta variant likely entered the country via the northern border provinces. CONCLUSIONS Continual genomic surveillance and sequencing efforts, in combination with public health strategies, play a vital role in effectively tracking and responding to the emergence, evolution and dissemination of future emerging variants.
Collapse
Affiliation(s)
- Yvonne C F Su
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore.
| | - Michael A Zeller
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Tey Putita Ou
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
- Ecole Doctorale GAIA, University of Montpelier, Montpelier, France
| | - Jordan Ma
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Leakhena Pum
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Rong Zhang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Sophannadeth Rath
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Vireak Heang
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sonita Kol
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Reaksa Lim
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Kim Lay Chea
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Limmey Khun
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Leangyi Heng
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sidonn Krang
- Communicable Disease Control Department, Ministry of Health, Phnom Penh, Cambodia
| | | | - Michael H Kinzer
- United States Centers for Disease Control and Prevention, Phnom Penh, Cambodia
| | - Vanra Ieng
- World Health Organization Country Office, Phnom Penh, Cambodia
| | - Vannda Kab
- World Health Organization Country Office, Phnom Penh, Cambodia
| | - Sarika Patel
- World Health Organization Country Office, Phnom Penh, Cambodia
| | - Borann Sar
- United States Centers for Disease Control and Prevention, Phnom Penh, Cambodia
| | - Viseth Srey Horm
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sokhoun Yann
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Heidi Auerswald
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Jurre Y Siegers
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Cecile Troupin
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
- Medical Virology and Rabies group, Institut Pasteur du Laos, Vientiane, Lao PDR, Laos
| | - Narjis Boukli
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Koen Vandelannoote
- Bacterial Phylogenomics Group, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Foong Ying Wong
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Giselle G K Ng
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Malen Chan
- Epidemiology and Public Health Unit, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sopheak Sorn
- Epidemiology and Public Health Unit, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Yi Sengdoeurn
- Communicable Disease Control Department, Ministry of Health, Phnom Penh, Cambodia
| | - Seng Heng
- Communicable Disease Control Department, Ministry of Health, Phnom Penh, Cambodia
| | - Chau Darapheak
- National Institute for Public Health, Phnom Penh, Cambodia
| | - Chin Savuth
- National Institute for Public Health, Phnom Penh, Cambodia
| | | | - Sowath Ly
- Epidemiology and Public Health Unit, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Laurence Baril
- Direction, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Andre Spiegel
- Direction, Institute Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Veasna Duong
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sovann Ly
- Communicable Disease Control Department, Ministry of Health, Phnom Penh, Cambodia
| | - Gavin J D Smith
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
- Centre for Outbreak Preparedness, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Erik A Karlsson
- Virology Unit, World Health Organization COVID-19 Global Referral Laboratory, Institut Pasteur du Cambodge, Phnom Penh, Cambodia.
| |
Collapse
|
45
|
Gkekas I, Katsamakas S, Mylonas S, Fotopoulou T, Magoulas GΕ, Tenchiu AC, Dimitriou M, Axenopoulos A, Rossopoulou N, Kostova S, Wanker EE, Katsila T, Papahatjis D, Gorgoulis VG, Koufaki M, Karakasiliotis I, Calogeropoulou T, Daras P, Petrakis S. AI Promoted Virtual Screening, Structure-Based Hit Optimization, and Synthesis of Novel COVID-19 S-RBD Domain Inhibitors. J Chem Inf Model 2024; 64:8562-8585. [PMID: 39535926 PMCID: PMC11600510 DOI: 10.1021/acs.jcim.4c01110] [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: 07/01/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by a new, highly pathogenic severe-acute-respiratory syndrome coronavirus 2 (SARS-CoV-2) that infects human cells through its transmembrane spike (S) glycoprotein. The receptor-binding domain (RBD) of the S protein interacts with the angiotensin-converting enzyme II (ACE2) receptor of the host cells. Therefore, pharmacological targeting of this interaction might prevent infection or spread of the virus. Here, we performed a virtual screening to identify small molecules that block S-ACE2 interaction. Large compound libraries were filtered for drug-like properties, promiscuity and protein-protein interaction-targeting ability based on their ADME-Tox descriptors and also to exclude pan-assay interfering compounds. A properly designed AI-based virtual screening pipeline was applied to the remaining compounds, comprising approximately 10% of the starting data sets, searching for molecules that could bind to the RBD of the S protein. All molecules were sorted according to their screening score, grouped based on their structure and postfiltered for possible interaction patterns with the ACE2 receptor, yielding 31 hits. These hit molecules were further tested for their inhibitory effect on Spike RBD/ACE2 (19-615) interaction. Six compounds inhibited the S-ACE2 interaction in a dose-dependent manner while two of them also prevented infection of human cells from a pseudotyped virus whose entry is mediated by the S protein of SARS-CoV-2. Of the two compounds, the benzimidazole derivative CKP-22 protected Vero E6 cells from infection with SARS-CoV-2, as well. Subsequent, hit-to-lead optimization of CKP-22 was effected through the synthesis of 29 new derivatives of which compound CKP-25 suppressed the Spike RBD/ACE2 (19-615) interaction, reduced the cytopathic effect of SARS-CoV-2 in Vero E6 cells (IC50 = 3.5 μM) and reduced the viral load in cell culture supernatants. Early in vitro ADME-Tox studies showed that CKP-25 does not possess biodegradation or liver metabolism issues, while isozyme-specific CYP450 experiments revealed that CKP-25 was a weak inhibitor of the CYP450 system. Moreover, CKP-25 does not elicit mutagenic effect on Escherichia coli WP2 uvrA strain. Thus, CKP-25 is considered a lead compound against COVID-19 infection.
Collapse
Affiliation(s)
- Ioannis Gkekas
- Institute
of Applied Biosciences, Centre for Research
and Technology Hellas, Thessaloniki 57001, Greece
| | - Sotirios Katsamakas
- Information
Technologies Institute, Centre for Research
and Technology Hellas, Thessaloniki 57001, Greece
- Institute
of Chemical Biology, National Hellenic Research
Foundation, 48 Vassileos Constantinou Avenue, Athens 11635, Greece
| | - Stelios Mylonas
- Information
Technologies Institute, Centre for Research
and Technology Hellas, Thessaloniki 57001, Greece
| | - Theano Fotopoulou
- Institute
of Chemical Biology, National Hellenic Research
Foundation, 48 Vassileos Constantinou Avenue, Athens 11635, Greece
| | - George Ε. Magoulas
- Institute
of Chemical Biology, National Hellenic Research
Foundation, 48 Vassileos Constantinou Avenue, Athens 11635, Greece
| | - Alia Cristina Tenchiu
- Institute
of Chemical Biology, National Hellenic Research
Foundation, 48 Vassileos Constantinou Avenue, Athens 11635, Greece
| | - Marios Dimitriou
- Laboratory
of Biology, Department of Medicine, Democritus
University of Thrace, Alexandroupolis 68100, Greece
| | - Apostolos Axenopoulos
- Information
Technologies Institute, Centre for Research
and Technology Hellas, Thessaloniki 57001, Greece
| | - Nafsika Rossopoulou
- Institute
of Chemical Biology, National Hellenic Research
Foundation, 48 Vassileos Constantinou Avenue, Athens 11635, Greece
| | - Simona Kostova
- Max-Delbrueck-Center
for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Erich E. Wanker
- Max-Delbrueck-Center
for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Theodora Katsila
- Institute
of Chemical Biology, National Hellenic Research
Foundation, 48 Vassileos Constantinou Avenue, Athens 11635, Greece
| | - Demetris Papahatjis
- Institute
of Chemical Biology, National Hellenic Research
Foundation, 48 Vassileos Constantinou Avenue, Athens 11635, Greece
| | - Vassilis G. Gorgoulis
- Molecular
Carcinogenesis Group, Department of Histology and Embryology, Medical
School, National and Kapodistrian University
of Athens, Athens 11635, Greece
- Ninewells
Hospital and Medical School, University
of Dundee, DD19SY Dundee, U.K.
| | - Maria Koufaki
- Institute
of Chemical Biology, National Hellenic Research
Foundation, 48 Vassileos Constantinou Avenue, Athens 11635, Greece
| | - Ioannis Karakasiliotis
- Laboratory
of Biology, Department of Medicine, Democritus
University of Thrace, Alexandroupolis 68100, Greece
| | - Theodora Calogeropoulou
- Institute
of Chemical Biology, National Hellenic Research
Foundation, 48 Vassileos Constantinou Avenue, Athens 11635, Greece
| | - Petros Daras
- Information
Technologies Institute, Centre for Research
and Technology Hellas, Thessaloniki 57001, Greece
| | - Spyros Petrakis
- Institute
of Applied Biosciences, Centre for Research
and Technology Hellas, Thessaloniki 57001, Greece
| |
Collapse
|
46
|
Lu Y, Yang Q, Ran T, Zhang G, Li W, Zhou P, Tang J, Dai M, Zhong J, Chen H, He P, Zhou A, Xue B, Chen J, Zhang J, Yang S, Wu K, Wu X, Tang M, Zhang WK, Guo D, Chen X, Chen H, Shang J. Discovery of orally bioavailable SARS-CoV-2 papain-like protease inhibitor as a potential treatment for COVID-19. Nat Commun 2024; 15:10169. [PMID: 39580525 PMCID: PMC11585628 DOI: 10.1038/s41467-024-54462-0] [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: 05/24/2024] [Accepted: 11/07/2024] [Indexed: 11/25/2024] Open
Abstract
The RNA-dependent RNA polymerase (RdRp), 3C-like protease (3CLpro), and papain-like protease (PLpro) are pivotal components in the viral life cycle of SARS-CoV-2, presenting as promising therapeutic targets. Currently, all FDA-approved antiviral drugs against SARS-CoV-2 are RdRp or 3CLpro inhibitors. However, the mutations causing drug resistance have been observed in RdRp and 3CLpro from SARS-CoV-2, which makes it necessary to develop antivirals with novel mechanisms. Through the application of a structure-based drug design (SBDD) approach, we discover a series of novel potent non-covalent PLpro inhibitors with remarkable in vitro potency and in vivo PK properties. The co-crystal structures of PLpro with lead compounds reveal that the residues D164 and Q269 around the S2 site are critical for improving the inhibitor's potency. The lead compound GZNL-P36 not only inhibits SARS-CoV-2 and its variants at the cellular level with EC50 ranging from 58.2 nM to 306.2 nM, but also inhibits HCoV-NL63 and HCoV-229E with EC50 of 81.6 nM and 2.66 μM, respectively. Oral administration of the GZNL-P36 results in significantly improved survival and notable reductions in lung viral loads and lesions in SARS-CoV-2 infection mouse model, consistent with RNA-seq data analysis. Our results indicate that PLpro inhibitors represent a promising SARS-CoV-2 therapy.
Collapse
Affiliation(s)
- Yongzhi Lu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- School of Basic Medical Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qi Yang
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ting Ran
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Guihua Zhang
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Wenqi Li
- Guangzhou National Laboratory, Guangzhou, 510005, China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Peiqi Zhou
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Jielin Tang
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Minxian Dai
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Jinpeng Zhong
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Hua Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Pan He
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Anqi Zhou
- School of Basic Medical Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, 511436, China
| | - Bao Xue
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Jiayi Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Jiyun Zhang
- Guangzhou National Laboratory, Guangzhou, 510005, China
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Sidi Yang
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Kunzhong Wu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xinyu Wu
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Miru Tang
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Wei K Zhang
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Deyin Guo
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Hongming Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- School of Basic Medical Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Jinsai Shang
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- School of Basic Medical Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
47
|
Wilasang C, Suttirat P, Wannigama DL, Amarasiri M, Chadsuthi S, Modchang C. Impact of COVID-19 Vaccination in Thailand: Averted Deaths and Severe Infections Across Age Groups. Trop Med Infect Dis 2024; 9:286. [PMID: 39728813 DOI: 10.3390/tropicalmed9120286] [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: 09/17/2024] [Revised: 11/12/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
The COVID-19 pandemic has underscored the pivotal role of vaccines in mitigating the devastating impact of the virus. In Thailand, the vaccination campaign against SARS-CoV-2 began on 28 February 2021, initially prioritizing healthcare professionals before expanding into a nationwide effort on 7 June 2021. This study employs a mathematical model of COVID-19 transmission with vaccination to analyze the impact of Thailand's COVID-19 vaccination program from 1 March 2021 to 31 December 2022. We specifically assess the potential loss of lives and occurrence of severe infections across various age groups in a hypothetical scenario where vaccines were not administered. By fitting our model with officially reported COVID-19 death data, our analysis reveals that vaccination efforts prevented a total of 300,234 deaths (95% confidence interval: 295,938-304,349) and averted 1.60 million severe COVID-19 infections (95% confidence interval: 1.54-1.65 million). Notably, the elderly population over 80 years old benefited the most from vaccination, with an estimated 84,518 lives saved, constituting 4.28% of this age group. Furthermore, individuals aged between 70 and 74 years experienced the highest reduction in severe infections, with vaccination potentially preventing 8.35% of this age bracket from developing severe COVID-19.
Collapse
Affiliation(s)
- Chaiwat Wilasang
- Biophysics Group, Department of Physics, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Center for Disease Modeling, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Pikkanet Suttirat
- Biophysics Group, Department of Physics, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Center for Disease Modeling, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Dhammika Leshan Wannigama
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata 990-2292, Japan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10400, Thailand
- Center of Excellence in Antimicrobial Resistance and Stewardship, Faculty of Medicine, Chulalongkorn University, Bangkok 10400, Thailand
- School of Medicine, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, Western Australia, Perth, WA 6009, Australia
- Biofilms and Antimicrobial Resistance Consortium of ODA Receiving Countries, The University of Sheffield, Sheffield S10 2TN, UK
- Pathogen Hunter's Research Collaborative Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata 990-2292, Japan
| | - Mohan Amarasiri
- Environmental Water Quality Engineering Laboratory, Department of Civil and Environmental Engineer-ing, Graduate School of Engineering, Tohoku University, 6-6-06, Aramaki, Aoba-ku, Sendai 980-8579, Japan
| | - Sudarat Chadsuthi
- Department of Physics, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Charin Modchang
- Biophysics Group, Department of Physics, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Center for Disease Modeling, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Centre of Excellence in Mathematics, Ministry of Higher Education, Science, Research and Innovation, Bangkok 10400, Thailand
- Thailand Center of Excellence in Physics, Ministry of Higher Education, Science, Research and Innovation, 328 Si Ayutthaya Road, Bangkok 10400, Thailand
| |
Collapse
|
48
|
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
|
49
|
Wang X, Gotchev D, Fan KY, Vega MM, Mani N, McGovern-Gooch K, Cuconati A, Tercero B, Wang X, Carpino P, Maskos K, Centrella PA, Schmitt A, Preuss F, Prasad A, Chen CY, Clark MA, Guilinger JP, Johnstone S, von König K, Keefe AD, Liu J, Turcotte S, Zhang Y, Konz Makino DL, Lam AM, Cole AG, Sofia MJ. Rational Design of Macrocyclic Noncovalent Inhibitors of SARS-CoV-2 M pro from a DNA-Encoded Chemical Library Screening Hit That Demonstrate Potent Inhibition against Pan-Coronavirus Homologues and Nirmatrelvir-Resistant Variants. J Med Chem 2024; 67:19623-19667. [PMID: 39453309 DOI: 10.1021/acs.jmedchem.4c02009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
The recent global COVID-19 pandemic has highlighted treatments for coronavirus infection as an unmet medical need. The main protease (Mpro) has been an important target for the development of SARS-CoV-2 direct-acting antivirals. Nirmatrelvir as a covalent Mpro inhibitor was the first such approved therapy. Although Mpro inhibitors of various chemical classes have been reported, they are generally less active against nirmatrelvir-resistant variants and have limited pan-coronavirus potential, presenting a significant human health risk upon future outbreaks. We here present a novel approach and utilized DNA-encoded chemical library screening to identify the noncovalent Mpro inhibitor 5, which demonstrated a distinct binding mode to nirmatrelvir. A macrocyclization strategy designed to lock the active conformation resulted in lactone 12 with significantly improved antiviral activity. Further optimization led to the potent lactam 26, which demonstrated exceptional potency against nirmatrelvir-resistant variants as well as against a panel of viral main proteases from other coronaviruses.
Collapse
Affiliation(s)
- Xu Wang
- Arbutus Biopharma Inc., 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Dimitar Gotchev
- Arbutus Biopharma Inc., 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Kristi Yi Fan
- Arbutus Biopharma Inc., 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Marvin M Vega
- Arbutus Biopharma Inc., 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Nagraj Mani
- Arbutus Biopharma Inc., 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | | | - Andrea Cuconati
- Arbutus Biopharma Inc., 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Breanna Tercero
- Arbutus Biopharma Inc., 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Xiaohe Wang
- Arbutus Biopharma Inc., 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Philip Carpino
- X-Chem Inc., 4800 Rue Levy Suite 200, Montreal, QC CA H4R 2P7, Canada
| | - Klaus Maskos
- Proteros Biostructures GmbH, Bunsenstraße 7a, Martinsried, Bavaria 82152, Germany
| | - Paolo A Centrella
- X-Chem Inc., 100 Beaver Street Suite 101, Waltham, Massachusetts 02453, United States
| | - Andreas Schmitt
- Proteros Biostructures GmbH, Bunsenstraße 7a, Martinsried, Bavaria 82152, Germany
| | - Franziska Preuss
- Proteros Biostructures GmbH, Bunsenstraße 7a, Martinsried, Bavaria 82152, Germany
| | - Archna Prasad
- Proteros Biostructures GmbH, Bunsenstraße 7a, Martinsried, Bavaria 82152, Germany
| | - Chia-Yi Chen
- Proteros Biostructures GmbH, Bunsenstraße 7a, Martinsried, Bavaria 82152, Germany
| | - Matthew A Clark
- X-Chem Inc., 100 Beaver Street Suite 101, Waltham, Massachusetts 02453, United States
| | - John P Guilinger
- X-Chem Inc., 100 Beaver Street Suite 101, Waltham, Massachusetts 02453, United States
| | - Shawn Johnstone
- X-Chem Inc., 4800 Rue Levy Suite 200, Montreal, QC CA H4R 2P7, Canada
| | - Konstanze von König
- Proteros Biostructures GmbH, Bunsenstraße 7a, Martinsried, Bavaria 82152, Germany
| | - Anthony D Keefe
- X-Chem Inc., 100 Beaver Street Suite 101, Waltham, Massachusetts 02453, United States
| | - Jenny Liu
- X-Chem Inc., 100 Beaver Street Suite 101, Waltham, Massachusetts 02453, United States
| | - Stéphane Turcotte
- X-Chem Inc., 4800 Rue Levy Suite 200, Montreal, QC CA H4R 2P7, Canada
| | - Ying Zhang
- X-Chem Inc., 100 Beaver Street Suite 101, Waltham, Massachusetts 02453, United States
| | - Debora L Konz Makino
- Proteros Biostructures GmbH, Bunsenstraße 7a, Martinsried, Bavaria 82152, Germany
| | - Angela M Lam
- Arbutus Biopharma Inc., 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Andrew G Cole
- Arbutus Biopharma Inc., 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Michael J Sofia
- Arbutus Biopharma Inc., 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| |
Collapse
|
50
|
Maher S, Assaly NME, Aly DM, Atta S, Fteah AM, Badawi H, Zahran MY, Kamel M. Comparative study of neutralizing antibodies titers in response to different types of COVID-19 vaccines among a group of egyptian healthcare workers. Virol J 2024; 21:277. [PMID: 39501293 PMCID: PMC11539826 DOI: 10.1186/s12985-024-02546-0] [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: 03/15/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Defining the protective thresholds against the severe-acute-respiratory-syndrome-related corona virus-2 pandemic is a crucial challenge. To reduce the risks of severe disease, hospitalization, and death, various COVID-19 vaccines have been rapidly developed. AIM OF THE WORK This study aimed to assess the impact of three common COVID-19 vaccine types; two mRNA COVID-19 vaccines: (Pfizer/BioNTech's BNT162b2 and Moderna's mRNA-1273), one adenoviral vector vaccine: Oxford/AstraZeneca's ChAdOx1, and one inactivated vaccine (Sinovac Biotech/China's Sinovac) on the level of neutralizing antibodies, considering factors such as vaccine type, demographic characteristics, and hybrid immunity. We conducted a direct comparative analysis involving 300 healthcare workers, both with and without prior SARS-CoV-2 infection (B.1, C.36.3, and AY.32 (Delta) variants). Neutralizing antibodies levels were measured at baseline (before vaccination), before the second dose, and six months after the second dose. RESULTS The results showed a significant increase in neutralizing antibodies levels after complete vaccination with all vaccine types. Among healthcare workers, those vaccinated with mRNA vaccines (Moderna or Pfizer) exhibited the highest neutralizing antibodies titers, followed by AstraZeneca, and finally Sinovac with the lowest titer. On studying the effect of previous COVID-19 infection after vaccination, no significant difference in neutralizing antibodies levels was observed between healthcare workers vaccinated with mRNA or AstraZeneca vaccines, both with prior COVID-19 infection, following the first and six months after the second dose. CONCLUSION These findings suggest that individuals with prior COVID-19 may only require a single dose of mRNA or AstraZeneca vaccines to achieve a similar level of immunization as those without prior COVID-19 who completed the vaccination program. HIGHLIGHTS There is a significant increase in neutralizing antibodies levels after complete vaccination against COVID-19 Vaccination with mRNA vaccines exhibits the highest neutralizing antibodies titers. Vaccination with Sinovac exhibits the lowest neutralizing antibodies titers.
Collapse
Affiliation(s)
- Sara Maher
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Nihal M El Assaly
- Clinical Chemistry Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Doaa Mamdouh Aly
- Clinical Chemistry Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Shimaa Atta
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt.
| | - Asmaa Mohamed Fteah
- Clinical Chemistry Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Hala Badawi
- Microbiology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | | | - Manal Kamel
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| |
Collapse
|