1
|
Hwang YH, Min DH, Beom Park W. Limitations of neutralizing antibody titers in COVID-19 vaccine efficacy trials and a call for additional correlates of protection. Hum Vaccin Immunother 2025; 21:2473795. [PMID: 40051347 PMCID: PMC11901426 DOI: 10.1080/21645515.2025.2473795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The coronavirus disease (COVID-19) pandemic accelerated development of various vaccine platforms. Among them, mRNA vaccines played a crucial role in controlling the pandemic due to their swift development and efficacy against virus variants. Despite the success of these vaccines, recent studies highlight challenges in evaluating vaccine efficacy, especially in individuals with prior COVID-19 infection. Weakened neutralizing antibody responses after additional doses are observed in these populations, raising concerns about using neutralizing antibody titers as the sole immune correlate of protection. While neutralizing antibodies remain the primary endpoint in immunogenicity trials, they may not fully capture the immune response in populations with widespread prior infection or vaccination. This review explores reduced neutralizing antibody responses in previously infected individuals, and their impact on vaccine efficacy evaluation. It also offers recommendations for improving efficacy assessment, stressing incorporation of additional immune markers such as cell-mediated immunity to enable more comprehensive understanding of vaccine-induced immunity.
Collapse
Affiliation(s)
- Young Hoon Hwang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dal-Hee Min
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Wan Beom Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
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
|
Glaser N, Diexer S, Klee B, Massag J, Pfrommer LR, Purschke O, Binder M, Frese T, Girndt M, Hoell JI, Moor I, Rosendahl J, Gekle M, Sedding D, Gottschick C, Mikolajczyk R. Duration of mild acute SARS-CoV-2 infections with Omicron depending on previous vaccinations and infections - Using data of the German DigiHero cohort study from post-pandemic winters 2022/2023 and 2023/2024. J Infect Public Health 2025; 18:102746. [PMID: 40090170 DOI: 10.1016/j.jiph.2025.102746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/23/2025] [Accepted: 03/09/2025] [Indexed: 03/18/2025] Open
Abstract
OBJECTIVES Protection against severe course of SARS-CoV-2 infection after COVID-19 vaccination or infection was extensively studied. It is unknown whether this effect also translates into shortened duration of mild infections. We assessed the duration of symptoms depending on vaccination status and previous SARS-CoV-2 infections among individuals with a mild course of infection. METHODS For two post-pandemic winters (2022/2023 and 2023/2024), in total 13,615 participants of the German DigiHero study reported their SARS-CoV-2 infections from September to March. Via negative binomial regression adjusting for sociodemographic factors, we studied the association of infection duration (days with symptoms and in bed) with number of vaccinations, prior SARS-CoV-2 infections, and time since last vaccination/and infection. RESULTS We noted no major differences in infection duration depending on the number of vaccinations and time since last infection for short mild infections (≤21 days with symptoms). Per 6 months since the last vaccination, symptom duration and days spent in bed increased by 2 % and 4 %. The risk of long mild SARS-CoV-2 infections (>21 days with symptoms) was higher for individuals with no prior SARS-CoV-2 infection (Odds Ratio: 1.98; 95 % confidence interval [1.43; 2.76]), but not for vaccinations (OR: 0.98; 95 % CI [0.74; 1.33]). CONCLUSIONS There was no indication of reduced duration of symptoms during short mild infections depending on the number of vaccinations and time since the last SARS-CoV-2 vaccination or infection. A prior SARS-CoV-2 infection was protective against prolonged disease in mild SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Nadine Glaser
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Centre for Health Sciences, Medical Faculty of the Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle 06112, Germany
| | - Sophie Diexer
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Centre for Health Sciences, Medical Faculty of the Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle 06112, Germany
| | - Bianca Klee
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Centre for Health Sciences, Medical Faculty of the Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle 06112, Germany
| | - Janka Massag
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Centre for Health Sciences, Medical Faculty of the Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle 06112, Germany
| | - Laura R Pfrommer
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Centre for Health Sciences, Medical Faculty of the Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle 06112, Germany
| | - Oliver Purschke
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Centre for Health Sciences, Medical Faculty of the Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle 06112, Germany
| | - Mascha Binder
- Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, Halle 06120, Germany; Medical Oncology, University Hospital Basel, Petersgraben 4, Basel 4031, Switzerland
| | - Thomas Frese
- Institute of General Practice and Family Medicine, Interdisciplinary Centre for Health Sciences, Medical Faculty of the Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle 06112, Germany
| | - Matthias Girndt
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, Halle 06120, Germany
| | - Jessica I Hoell
- Paediatric Haematology and Oncology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, Halle 06120, Germany
| | - Irene Moor
- Institute for Medical Sociology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle 06112, Germany
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, Halle 06120, Germany
| | - Michael Gekle
- Julius-Bernstein-Institute of Physiology, Medical Faculty of the Martin Luther University, Halle-Wittenberg, Magdeburger Straße 6, Halle 06112, Germany
| | - Daniel Sedding
- Mid-German Heart Centre, Department of Cardiology and Intensive Care Medicine, University Hospital, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, Halle 06120, Germany
| | - Cornelia Gottschick
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Centre for Health Sciences, Medical Faculty of the Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle 06112, Germany
| | - Rafael Mikolajczyk
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Centre for Health Sciences, Medical Faculty of the Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle 06112, Germany.
| |
Collapse
|
4
|
Yasmin S, Ansari MY. A detailed examination of coronavirus disease 2019 (COVID-19): Covering past and future perspectives. Microb Pathog 2025; 203:107398. [PMID: 39986548 DOI: 10.1016/j.micpath.2025.107398] [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: 07/06/2024] [Revised: 01/07/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
The COVID-19 disease has spread rapidly across the world within just six months, affecting 169 million people and causing 3.5 million deaths globally (2021). The most affected countries include the USA, Brazil, India, and several European countries such as the UK and Russia. Healthcare professionals face new challenges in finding better ways to manage patients and save lives. In this regard, more comprehensive research is needed, including genomic and proteomic studies, personalized medicines and the design of suitable treatments. However, finding novel molecular entities (NME) using a standard or de novo strategy to drug development is a time-consuming and costly process. Another alternate strategy is discovering new therapeutic uses for old/existing/available medications, known as drug repurposing. There are a variety of computational repurposing methodologies, and some of them have been used to counter the coronavirus disease pandemic of 2019 (COVID-19). This review article compiles recently published data on the origin, transmission, pathogenesis, diagnosis, and management of the coronavirus by drug repurposing and vaccine development approach. We have attempted to screen probable drugs in clinical trials by using literature survey. This systematic review aims to create priorities for future research of drugs repurposed and vaccine development for COVID-19.
Collapse
Affiliation(s)
- Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133207, India; Ibne Seena College of Pharmacy, Azmi Vidya Nagri Anjhi Shahabad, Hardoi, Uttar Pradesh (U.P.) 241124, India.
| |
Collapse
|
5
|
Aga AM, Mulugeta D, Gebreegziabxier A, Zeleke GT, Girmay AM, Tura GB, Ayele A, Mohammed A, Belete T, Taddele T, Abubeker R, Woldemariyam FT, Gelanew T, Tesera Y, Gidisa B, Tura JB, Leta GT, Ali A, Beshah SA, Likasa BW, Mohammed J, Nigussie D. Genome diversity of SARS-CoV-2 lineages associated with vaccination breakthrough infections in Addis Ababa, Ethiopia. BMC Infect Dis 2025; 25:738. [PMID: 40410660 DOI: 10.1186/s12879-025-11107-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: 10/16/2024] [Accepted: 05/12/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Extensive vaccination campaigns against COVID-19 have played a significant role in controlling virus spread and preventing severe illness. This study focused on breakthrough infections in vaccinated individuals, raising concerns about vaccine effectiveness against SARS-CoV-2 variant immune escape, with particular attention to lineage distribution among vaccinated and unvaccinated individuals. METHODS A case-control study was conducted from January to April 2023, sequencing 298 samples from participants who tested positive for COVID-19 via rapid diagnostic test (RDT) from 22 health facilities, including vaccinated and unvaccinated cases. Besides clinical and epidemiological data, nasopharyngeal swabs were obtained, and reverse transcription quantitative polymerase chain reaction (RT-qPCR) was conducted to determine Cycle threshold (Ct) values, followed by whole genome sequencing of 298 samples fulfilling sequencing criteria to identify variants of concern and specific virus lineages. RESULTS Out of 298 samples sequenced, 281 fulfill quality for analysis with 44.8% (126) had received at least one COVID-19 vaccine dose, while 51.9% (146) were not vaccinated, and 3.2% (9) patients had no vaccination records. The analysis showed that all cases were of the Omicron variant, with the XBB.1.5 lineage being the most prevalent (38.4%), followed by FL.2 (9.3%) and XBB.1.9.1.2 (7.8%). The remaining 44.5% comprised a combination of 22 other lineages. The XBB.1.5 variant accounted for 51 (47.2%) cases among vaccinated individuals with at least one dose and 57 (52.8%) among unvaccinated, showing relatively similar prevalence across both groups. The viral load as indicated by the Ct value varied widely, with a significant appearance in the lower ranges (high viral load), suggesting active viral replication. Notably, 25% of samples exhibited high viral loads (Ct values 13-15), showing the high transmissibility of the XBB.1.5 lineage among both vaccinated and unvaccinated populations. CONCLUSION The findings emphasize the need for continuous genomic surveillance and regular vaccine updates to address emerging SARS-CoV-2 variants, particularly the immune-evasive XBB lineage. The high prevalence of variants like XBB.1.5 in breakthrough infection underscores the importance of adaptive vaccination strategies and next-generation vaccines to maintain efficacy. Ongoing monitoring of variant dynamics is crucial for informed public health responses, strengthening pandemic preparedness and future outbreak prevention.
Collapse
Affiliation(s)
- Abebe M Aga
- Armauer Hansen Research Institute, Addis Ababa, P.O Box 1005, Ethiopia.
| | - Demise Mulugeta
- Armauer Hansen Research Institute, Addis Ababa, P.O Box 1005, Ethiopia
| | | | - Girum Taye Zeleke
- Ethiopian Public Health Institute, P.O Box 1242, Addis Ababa, Ethiopia
| | | | - Gutema Bulti Tura
- Ethiopian Public Health Institute, P.O Box 1242, Addis Ababa, Ethiopia
| | - Abaysew Ayele
- Armauer Hansen Research Institute, Addis Ababa, P.O Box 1005, Ethiopia
| | - Ahmed Mohammed
- Ethiopian Public Health Institute, P.O Box 1242, Addis Ababa, Ethiopia
| | - Tigist Belete
- Ethiopian Public Health Institute, P.O Box 1242, Addis Ababa, Ethiopia
| | - Tefera Taddele
- Ethiopian Public Health Institute, P.O Box 1242, Addis Ababa, Ethiopia
| | - Rajiha Abubeker
- Ethiopian Public Health Institute, P.O Box 1242, Addis Ababa, Ethiopia
| | | | - Tesfaye Gelanew
- Armauer Hansen Research Institute, Addis Ababa, P.O Box 1005, Ethiopia
| | - Yeweynshet Tesera
- Armauer Hansen Research Institute, Addis Ababa, P.O Box 1005, Ethiopia
| | - Bedasa Gidisa
- Armauer Hansen Research Institute, Addis Ababa, P.O Box 1005, Ethiopia
| | - Jaleta Bulti Tura
- Ethiopian Public Health Institute, P.O Box 1242, Addis Ababa, Ethiopia
| | | | - Abraham Ali
- Ethiopian Public Health Institute, P.O Box 1242, Addis Ababa, Ethiopia
| | | | | | - Jemal Mohammed
- Armauer Hansen Research Institute, Addis Ababa, P.O Box 1005, Ethiopia
| | - Dereje Nigussie
- Armauer Hansen Research Institute, Addis Ababa, P.O Box 1005, Ethiopia
| |
Collapse
|
6
|
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
|
7
|
Liu C, Tsang TK, Sullivan SG, Cowling BJ, Yang B. Comparative duration of neutralizing responses and protections of COVID-19 vaccination and correlates of protection. Nat Commun 2025; 16:4748. [PMID: 40404724 PMCID: PMC12098666 DOI: 10.1038/s41467-025-60024-9] [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: 08/22/2024] [Accepted: 05/06/2025] [Indexed: 05/24/2025] Open
Abstract
The decline in neutralizing antibody (nAb) titers and vaccine efficacy /effectiveness (VE) for SARS-CoV-2 vaccines has been observed over time and when confronted with emerging variants, two factors that are hard to distinguish. Despite substantial drop in nAb titers against Omicron, VE remains high for severe cases and fatalities, raising questions about the utility of detected nAbs as a correlate of protection for COVID-19 vaccines for varying disease severity. Here, we conducted a systematic comparison of waning dynamics of nAb and VE over time and against variants with varying levels of disease severity. Using Bayesian linear regression models, we found that antigenically-shifted variants, like Omicron, could potentially lead to greater reductions in nAb titers and primary VE against mild infections than associated immunity waning observed over a 180-day period. By comparing model predicted nAb titers and VE on the same time scales, we found that VE against severe and fatal outcomes remained above 75% even when nAb titers reached the detectable limit of assays, despite strong correlations with nAb titers (spearman correlations ≥0.7) across variants over time. This finding suggested detectable nAb titers are not always sensitive enough to fully predict protection against severe disease and death from SARS-CoV-2.
Collapse
Affiliation(s)
- Chang Liu
- World Health Organization 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, Hong Kong, China
| | - Tim K Tsang
- World Health Organization 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, Hong Kong, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, Hong Kong, China
| | - Sheena G Sullivan
- School of Clinical Sciences, Monash University, Melbourne, Australia
- Department of Epidemiology, University of California, Los Angeles, USA
| | - Benjamin J Cowling
- World Health Organization 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, Hong Kong, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, Hong Kong, China
| | - Bingyi Yang
- World Health Organization 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, Hong Kong, China.
| |
Collapse
|
8
|
Ward CL, Rojas Castro MY, Chakhunashvili G, Chitadze N, Finci I, Pebody R, Kissling E, Katz MA, Sanodze L. COVID-19 vaccine effectiveness among healthcare workers during the Omicron period in the country of Georgia, January - June 2022. PLoS One 2025; 20:e0311337. [PMID: 40397872 DOI: 10.1371/journal.pone.0311337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 04/14/2025] [Indexed: 05/23/2025] Open
Abstract
INTRODUCTION Understanding COVID-19 vaccine effectiveness (VE) in healthcare workers (HCWs) is critical to inform vaccination policies. We measured COVID-19 VE against laboratory-confirmed symptomatic infection in HCWs in the country of Georgia from January - June 2022, during a period of Omicron circulation. METHODS We conducted a cohort study of HCWs in six hospitals in Georgia. HCWs were enrolled in early 2021. Participants completed weekly symptom questionnaires. Symptomatic HCWs were tested by RT-PCR and/or rapid antigen test (RAT). Participants were also routinely tested, at varying frequencies during the study period, for SARS-CoV-2 by RT-PCR or RAT, regardless of symptoms. Serology was collected quarterly throughout the study and tested by electrochemiluminescence immunoassay for SARS-CoV-2 antibodies. We estimated absolute and relative VE of a first booster dose compared to a primary vaccine series as (1-hazard ratio)*100 using Cox proportional hazards models. RESULTS Among 1253 HCWs, 141 (11%) received a primary vaccine series (PVS) and a first booster, 855 (68%) received PVS only, and 248 (20%) were unvaccinated. Most boosters were BNT162b2 (Comirnaty original monovalent) vaccine (90%) and BBIBP-CorV vaccine (Sinopharm) (9%). Most PVS were BNT162b2 vaccine (68%) and BBIBP-CorV vaccine (24%). Absolute VE for a first booster was 40% (95% Confidence Interval (CI) -56-77) at 7-29 days following vaccination, -9% (95% CI -104-42) at 30-59 days following vaccination, and -46% (95% CI -156-17) at ≥ 60 days following vaccination. Relative VE of first booster dose compared to PVS was 58% (95% CI 1-82) at 7-29 days following vaccination, 21% (95% CI -33-54) at 30-59 days following vaccination, and -9% (95% CI -82-34) at ≥ 60 days following vaccination. CONCLUSION In Georgia, first booster dose VE against symptomatic SARS-CoV-2 infection among HCWs was moderately effective but waned very quickly during Omicron. Increased efforts to vaccinate priority groups in Georgia, such as healthcare workers, prior to periods of anticipated high COVID-19 incidence are essential.
Collapse
Affiliation(s)
| | | | | | | | - Iris Finci
- World Health Organization Regional Office for Europe, Copenhagen, Denmark
| | - Richard Pebody
- World Health Organization Regional Office for Europe, Copenhagen, Denmark
| | | | - Mark A Katz
- World Health Organization Regional Office for Europe, Copenhagen, Denmark
| | - Lia Sanodze
- National Center for Disease Control and Public Health, Tbilisi, Georgia
| |
Collapse
|
9
|
Schaffer AL, Hulme WJ, Horne E, Parker EPK, Walker V, Stables C, Mehrkar A, Bacon SCJ, Bates C, Goldacre B, Walker AJ, Hernán MA, Sterne JAC. Effect of the 2022 COVID-19 booster vaccination campaign in people aged 50 years in England: Regression discontinuity analysis in OpenSAFELY-TPP. Vaccine 2025; 59:127257. [PMID: 40398326 DOI: 10.1016/j.vaccine.2025.127257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 04/03/2025] [Accepted: 05/11/2025] [Indexed: 05/23/2025]
Abstract
SARS-CoV-2 vaccines are highly effective in preventing severe COVID-19 but require boosting to maintain protection. Changes to circulating variants and prevalent natural immunity may impact on real-world effectiveness of boosters. With NHS England approval, we used linked routine clinical data from >24 million patients to evaluate the effectiveness of the 2022 combined COVID-19 autumn booster and influenza vaccine campaign in non-clinically vulnerable 50-year-olds in England using a regression discontinuity design. Our primary outcome was a composite of 6-week COVID-19 emergency attendance, COVID-19 unplanned hospitalisation, or death. By 26 November 2022, booster vaccine coverage was 11.1 % at age 49.75 years increasing to 39.7 % at age 50.25 years. The estimated effect of the campaign on the risk of the primary outcome in 50-year-olds during weeks 7-12 after the start of the campaign was -0.4 per 100,000 (95 % CI -7.8, 7.1). The results were similar when using different follow-up start dates or when estimating the effect of vaccination (rather than the campaign). This study found little evidence that the autumn 2022 vaccination campaign in England was associated with a reduction in severe COVID-19-related outcomes among non-clinically vulnerable 50-year-olds. Possible explanations include the low risk of severe outcomes and substantial pre-existing vaccine- and infection-induced immunity. The booster campaign may have had effects beyond those we estimated, including reducing virus transmission and incidence of mild or moderate COVID-19.
Collapse
Affiliation(s)
- Andrea L Schaffer
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK.
| | - William J Hulme
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Elsie Horne
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK; National Institute of Health and Care Research, Bristol Biomedical Research Centre, Bristol, UK
| | - Edward P K Parker
- London School of Hygiene & Tropical Medicine, London, UK; National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Vaccines and Immunisation, London, UK
| | - Venexia Walker
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK; Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Catherine Stables
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Amir Mehrkar
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Seb C J Bacon
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | | | - Ben Goldacre
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Alex J Walker
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Miguel A Hernán
- CAUSALab, Harvard T.H. Chan School of Public Health, Boston, USA; Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Jonathan A C Sterne
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK; National Institute of Health and Care Research, Bristol Biomedical Research Centre, Bristol, UK; Health Data Research UK South-West, Bristol, UK
| |
Collapse
|
10
|
Narum S, Stalder T, Ridenhour B, Coats ER. SARS-CoV-2 surveillance of wastewater in small rural communities identifies lack of vaccine coverage as influence of omicron outbreak. WATER RESEARCH 2025; 283:123818. [PMID: 40381278 DOI: 10.1016/j.watres.2025.123818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 04/17/2025] [Accepted: 05/11/2025] [Indexed: 05/20/2025]
Abstract
Wastewater-based epidemiology (WBE) can provide critical early warnings to aid public health, which can be particularly beneficial in rural communities with limited access to health care. Spikes of SARS-CoV-2 RNA concentration in wastewater have been used to represent infections in a community, but wastewater holds a wealth of information that has not been explored yet. The objectives of this research were to expand the use of WBE to 1) determine the dynamic of SARS-CoV-2 variants in rural communities, and 2) evaluate the relationship between community vaccination status and the outbreak of a variant. We quantified the concentration of SARS-CoV-2 RNA, as well as specific mutations that are consistent with Delta and Omicron in influent raw wastewater samples collected from wastewater treatment facilities (WWTFs) for five populations with <1000 residents and one larger population in Latah County, ID. A binomial generalized linear model using the percent of the population with protection against Omicron from the initial vaccines and the booster shot was able to predict the probability of an uptick in Omicron concentration in wastewater with an accuracy of 0.96. Evaluation of vaccination data indicate that the spike in Omicron infections in December 2021 in the studied towns was linked to low levels of population protection from the initial shots of the COVID-19 vaccine against Omicron infection and limited uptake of booster shots in these communities. Despite difficulties with applying WBE in rural regions, this study shows that beyond evaluating spikes of viral infections, WBE can be used to evaluate the effect of a population's vaccine coverage on SARS-CoV-2 variant dynamics.
Collapse
Affiliation(s)
- Solana Narum
- Bioinformatics and Computational Biology Graduate Program (BCB), University of Idaho, 875 Perimeter Drive, MS 1103, Moscow, ID, 83844, United States
| | - Thibault Stalder
- Institute for Modeling Collaboration and Innovation (IMCI), University of Idaho, 875 Perimeter Drive, MS 1122, Moscow, ID, 83844, United States; Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, MS 3051, Moscow, ID, 83844, United States; Institute for Interdisciplinary Data Sciences (IIDS), University of Idaho, 875 Perimeter Drive, MS 3051, Moscow, ID, 83844, United States
| | - Benjamin Ridenhour
- Institute for Modeling Collaboration and Innovation (IMCI), University of Idaho, 875 Perimeter Drive, MS 1122, Moscow, ID, 83844, United States; Institute for Interdisciplinary Data Sciences (IIDS), University of Idaho, 875 Perimeter Drive, MS 3051, Moscow, ID, 83844, United States; Department of Mathematics and Statistical Science, University of Idaho, 875 Perimeter Drive, MS 1103, Moscow, ID, 83844, United States
| | - Erik R Coats
- Institute for Modeling Collaboration and Innovation (IMCI), University of Idaho, 875 Perimeter Drive, MS 1122, Moscow, ID, 83844, United States; Department of Civil and Environmental Engineering, University of Idaho, 875 Perimeter Drive, MS 1022, Moscow, ID, 83844, United States.
| |
Collapse
|
11
|
Hao T, Ryan GE, Lydeamore MJ, Cromer D, Wood JG, McVernon J, McCaw JM, Shearer FM, Golding N. Predicting immune protection against outcomes of infectious disease from population-level effectiveness data with application to COVID-19. Vaccine 2025; 55:126987. [PMID: 40117726 DOI: 10.1016/j.vaccine.2025.126987] [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: 11/01/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/23/2025]
Abstract
Quantifying the extent to which previous infections and vaccinations confer protection against future infection or disease outcomes is critical to managing the transmission and consequences of infectious diseases. We present a general statistical model for predicting the strength of protection conferred by different immunising exposures (numbers, types, and strains of both vaccines and infections), against multiple outcomes of interest, whilst accounting for immune waning. We predict immune protection against key clinical outcomes: developing symptoms, hospitalisation, and death. We also predict transmission-related outcomes: acquisition of infection and onward transmission in breakthrough infections. These enable quantification of the impact of immunity on population-level transmission dynamics. Our model calibrates the level of immune protection, drawing on both population-level data, such as vaccine effectiveness estimates, and neutralising antibody levels as a correlate of protection. This enables the model to learn realised immunity levels beyond those which can be predicted by antibody kinetics or other correlates alone. We demonstrate an application of the model for SARS-CoV-2, and predict the individual-level protective effectiveness conferred by natural infections with the Delta and the Omicron B.1.1.529 variants, and by the BioNTech-Pfizer (BNT162b2), Oxford-AstraZeneca (ChAdOx1), and 3rd-dose mRNA booster vaccines, against outcomes for both Delta and Omicron. We also demonstrate a use case of the model in late 2021 during the emergence of Omicron, showing how the model can be rapidly updated with emerging epidemiological data on multiple variants in the same population, to infer key immunogenicity and intrinsic transmissibility characteristics of the new variant, before the former can be more directly observed via vaccine effectiveness data. This model provided timely inference on rapidly evolving epidemic situations of significant concern during the early stages of the COVID-19 pandemic. The general nature of the model enables it to be used to support management of a range of infectious diseases.
Collapse
Affiliation(s)
- Tianxiao Hao
- The Kids Research Institute, Nedlands, Western Australia, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia.
| | - Gerard E Ryan
- The Kids Research Institute, Nedlands, Western Australia, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia
| | - Michael J Lydeamore
- Department of Econometrics and Business Statistics, Monash University, Victoria, Australia
| | - Deborah Cromer
- Kirby Institute, University of New South Wales Sydney, New South Wales, Australia
| | - James G Wood
- School of Population Health, University of New South Wales Sydney, New South Wales, Australia
| | - Jodie McVernon
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Victoria, Australia; Victorian Infectious Disease Reference Laboratory Epidemiology Unit, The Royal Melbourne Hospital, Victoria, Australia
| | - James M McCaw
- Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia; School of Mathematics and Statistics, The University of Melbourne, Victoria, Australia
| | - Freya M Shearer
- The Kids Research Institute, Nedlands, Western Australia, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia
| | - Nick Golding
- The Kids Research Institute, Nedlands, Western Australia, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia; School of Population Health, Curtin University, Western Australia, Australia
| |
Collapse
|
12
|
Jiang J, Lam KF, Lau EHY, Yin G, Lin Y, Cowling BJ. Estimation of trajectory of COVID-19 vaccines effectiveness against infection. Vaccine 2025; 55:127067. [PMID: 40158307 DOI: 10.1016/j.vaccine.2025.127067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025]
Abstract
This large-scale cohort study conducted in Hong Kong examined the time-varying protective effects of various COVID-19 vaccines and dosing regimens against the Omicron BA.1/BA.2 variants. An innovative pharmacokinetic/pharmacodynamic model was employed to estimate the trajectory of vaccine effectiveness over time. Results indicated that the maximum protection for a single dose reached 0.120 for CoronaVac and 0.171 for Comirnaty. The peak protective effectiveness for the second and third doses were observed at 0.348 and 0.522, respectively. In a 4-dose regimen, CoronaVac demonstrated a maximum protective effectiveness of 0.548, stabilizing at 0.487, while Comirnaty achieved a maximum effectiveness of 0.784, stabilizing at 0.714 six months after the administration of the last dose. The vaccine effectiveness exhibited a rising and then declining pattern, peaking approximately 1-2 months post-vaccination. Understanding waning immunity is crucial for optimizing vaccination strategies and policies as viral evolution continues. This real-world study captured changing dynamics that may differ from clinical trials with limited follow-up, providing essential evidence to guide the optimization of vaccination efforts. Ongoing monitoring of vaccine effectiveness remains critical as the viral landscape evolves. OBJECTIVES This study aims to investigate the time-varying protective effects of various COVID-19 vaccines and dosing regimens against infections caused by the Omicron BA.1/BA.2 in Hong Kong. METHODS This territory-wide cohort study from Hong Kong combined vaccination records, confirmed COVID-19 cases, and census data from January 2022 to May 2022 to comprehensively analyze the time-varying protective effects of different COVID-19 vaccines and dosing regimens against Omicron BA.1 and BA.2 infections. A 4-parameter pharmacokinetic/pharmacodynamic model was used to estimate the trajectory of vaccine effectiveness over time. RESULTS Among 6.2 million adults, the maximum protective effectiveness for a single vaccine dose reached 0.120 for CoronaVac and 0.171 for Comirnaty. For the second and third doses, peak effectiveness were observed at 0.348 for CoronaVac and 0.522 for Comirnaty. Notably, a 4-dose regimen resulted in maximum protections of 0.548 for CoronaVac and 0.785 for Comirnaty, which stabilized at 0.487 and 0.714, respectively, six months following the last doses. The vaccine effectiveness exhibited a rising then declining pattern, peaking around 1-2 months post-vaccination, underscoring the importance of ongoing vaccination strategies. CONCLUSIONS Understanding the waning of vaccine protection over time is critical for informing optimal vaccination strategies, booster schedules, and public health policies. This real-world study can capture changing dynamics that may differ from clinical trials which have more limited follow-up periods, and can provide crucial evidence to guide optimization of vaccination strategies. Ongoing monitoring of vaccine effectiveness remains crucial as the viral evolution continues.
Collapse
Affiliation(s)
- Jialiang Jiang
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of China
| | - Kwok Fai Lam
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of China; Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore.
| | - Eric Ho Yin Lau
- Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region of China
| | - Guosheng Yin
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of China
| | - Yun Lin
- Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region of China
| | - Benjamin John Cowling
- Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region of China; WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of China
| |
Collapse
|
13
|
Yang Q, Huang X, Zhang H, Sun J, Tang J, Chen Z, Liu L, Liu M, Sun Z, Tang Z, Wei D, Wang D, Wang Y, Yan M, Zhao L, Zhu A, Zhong Y, Yang H, Zhao Y, Dai J, Shi Y, Huang B, Zhang W, Zhao J, Chen X, Rao Z, Peng W. Expanding the utilization of binding pockets proves to be effective for noncovalent small molecule inhibitors against SARS-CoV-2 M pro. Eur J Med Chem 2025; 289:117497. [PMID: 40090296 DOI: 10.1016/j.ejmech.2025.117497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/27/2025] [Accepted: 03/08/2025] [Indexed: 03/18/2025]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in millions of deaths and continues to pose serious threats to global public health. The main protease (Mpro) of SARS-CoV-2 is crucial for viral replication and its conservation, making it an attractive drug target. Here, we employed a structure-based drug design strategy to develop and optimize novel inhibitors targeting SARS-CoV-2 Mpro. By fully exploring occupation of the S1, S2, and S3/S4 binding pockets, we identified eight promising inhibitors with half-maximal inhibitory concentration (IC50) values below 20 nM. The cocrystal structure of Mpro with compound 10 highlighted the crucial roles of the interactions within the S3/S4 pockets in inhibitor potency enhancement. These findings demonstrated that expanding the utilization of these binding pockets was an effective strategy for developing noncovalent small molecule inhibitors that target SARS-CoV-2 Mpro. Compound 4 demonstrated outstanding in vitro antiviral activity against wild-type SARS-CoV-2 with an EC50 of 9.4 nM. Moreover, oral treatment with compounds 1 and 9 exhibited excellent antiviral potency and substantially ameliorated virus-induced tissue damage in the lungs of Omicron BA.5-infected K18-human ACE2 (K18-hACE2) transgenic mice, indicating that these novel noncovalent inhibitors could be potential oral agents for the treatment of COVID-19.
Collapse
Affiliation(s)
- Qi Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Xupeng Huang
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Hongbo Zhang
- Beijing StoneWise Technology Co. Ltd., Beijing, 100080, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Jielin Tang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Zhao Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Lijie Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; Guangzhou Medical University, Guangzhou, 511436, China
| | - Man Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Zeyun Sun
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhenhao Tang
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Dandan Wei
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Dong Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Yiliang Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Mengrong Yan
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Li Zhao
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; Guangzhou Medical University, Guangzhou, 511436, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Yihang Zhong
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Haitao Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yao Zhao
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, 518112, China
| | - Jun Dai
- Technology Centre, Guangzhou Customs, Guangzhou, 510623, China
| | - Yongxia Shi
- Technology Centre, Guangzhou Customs, Guangzhou, 510623, China
| | - Bo Huang
- Beijing StoneWise Technology Co. Ltd., Beijing, 100080, China.
| | - Wei Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xinwen Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China.
| | - Zihe Rao
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; Laboratory of Structural Biology, School of Life Sciences and School of Medicine, Tsinghua University, Beijing, 100084, China; Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Wei Peng
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China; University of South China, Hengyang, 421001, China.
| |
Collapse
|
14
|
Costa SM, Simas MCDC, da Costa LJ, Silva R. Dynamics of SARS-CoV-2 Mutations in Wastewater Provide Insights into the Circulation of Virus Variants in the Population. Int J Mol Sci 2025; 26:4324. [PMID: 40362561 PMCID: PMC12072199 DOI: 10.3390/ijms26094324] [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: 03/14/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
SARS-CoV-2 high transmission and genomic mutations result in the emergence of new variants that impact COVID-19 vaccine efficacy and virus transmission by evading the host immune system. Wastewater-based epidemiology is an effective approach to monitor SARS-CoV-2 variants circulation in the population but is a challenge due to the presence of reaction inhibitors and the low concentrations of SARS-CoV-2 in this environment. Here, we aim to improve SARS-CoV-2 variant detection in wastewater by employing nested PCR followed by next-generation sequencing (NGS) of small amplicons of the S gene. Eight SARS-CoV-2 wastewater samples from Alegria Wastewater Treatment Plant, in Rio de Janeiro, Brazil, were collected monthly from February to September 2021. Samples were submitted to virus concentration, RNA extraction and nested PCR followed by NGS. The small amplicons were used to prepare libraries for sequencing without the need to perform any fragmentation step. We identified and calculated the frequencies of 29 mutations matching the Alpha, Beta, Gamma, Delta, Omicron, and P.2 variants. Omicron matching-mutations were detected before the lineage was classified as a variant of concern. SARS-CoV-2 wastewater sequences clustered with SARS-CoV-2 variants detected in clinical samples that circulated in 2021 in Rio de Janeiro. We show that sequencing of selected small amplicons of SARS-CoV-2 S gene allows the identification of SARS-CoV-2 variants matching mutations and their frequencies' calculation. This approach may be expanded using customizing primers for additional genomic regions, in order to differentiate current variants. Approaches that allow us to learn how variants emerge and how they relate to clinical outcomes are crucial for our understanding of the dynamics of virus variants circulation, providing valuable data for public health management.
Collapse
Affiliation(s)
- Sara Mesquita Costa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.M.C.); (M.C.d.C.S.)
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Maria Clara da Costa Simas
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.M.C.); (M.C.d.C.S.)
| | - Luciana Jesus da Costa
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Rosane Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.M.C.); (M.C.d.C.S.)
| |
Collapse
|
15
|
McGee JE, Kirsch JR, Kenney D, Cerbo F, Chavez EC, Shih TY, Douam F, Wong WW, Grinstaff MW. Complete substitution with modified nucleotides in self-amplifying RNA suppresses the interferon response and increases potency. Nat Biotechnol 2025; 43:720-726. [PMID: 38977924 PMCID: PMC11707045 DOI: 10.1038/s41587-024-02306-z] [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: 09/15/2023] [Accepted: 06/04/2024] [Indexed: 07/10/2024]
Abstract
The use of modified nucleotides to suppress the interferon response and maintain translation of self-amplifying RNA (saRNA), which has been achieved for mRNA, has not yet succeeded. We identify modified nucleotides that, when substituted at 100% in saRNA, confer innate immune evasion and robust long-term protein expression, and when formulated as a vaccine, protect against lethal SARS-CoV-2 challenge in mice. This discovery advances saRNA therapeutics by enabling prolonged protein expression at low doses.
Collapse
Affiliation(s)
- Joshua E McGee
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
| | - Jack R Kirsch
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Devin Kenney
- Department of Virology, Immunology and Microbiology, Boston University School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA
| | - Faith Cerbo
- Department of Virology, Immunology and Microbiology, Boston University School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA
| | - Elizabeth C Chavez
- Department of Virology, Immunology and Microbiology, Boston University School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA
| | - Ting-Yu Shih
- Department of Chemistry, Boston University, Boston, MA, USA
| | - Florian Douam
- Department of Virology, Immunology and Microbiology, Boston University School of Medicine, Boston, MA, USA.
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA.
| | - Wilson W Wong
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
- Biological Design Center, Boston University, Boston, MA, USA.
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
- Department of Chemistry, Boston University, Boston, MA, USA.
| |
Collapse
|
16
|
McCullough MD, Spencer BR, Shi J, Plumb ID, Haynes JM, Shah M, Briggs-Hagen M, Stramer SL, Jones JM, Midgley CM. Coronavirus Disease 2019 Symptoms by Immunity Status and Predominant-Variant Period Among US Blood Donors. Open Forum Infect Dis 2025; 12:ofaf185. [PMID: 40322271 PMCID: PMC12048779 DOI: 10.1093/ofid/ofaf185] [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: 11/22/2024] [Accepted: 03/24/2025] [Indexed: 05/08/2025] Open
Abstract
Background Amid changing variant and immunity landscapes since early in the coronavirus disease 2019 (COVID-19) pandemic, common COVID-19 symptoms need better understanding in relation to prior immunity or infecting variant. Methods American Red Cross blood donors were surveyed during February-April 2022 about prior COVID-19 vaccinations and symptomatic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. Donations were tested for anti-nucleocapsid antibodies to inform infection history. Restricting analysis to donors with survey-reported infections during the Omicron BA.1-predominant period (19 December 2021 through 19 March 2022), we used multivariable logistic regression to compare symptoms by existing immunity from prior infection or vaccination. Restricting analysis to those with no existing immunity, we compared symptoms by variant-predominant period of their first reported infection (BA.1 vs before). Results Among 9505 donors with a BA.1-predominant period infection, donors with prior infection (n = 1115), vaccination (n = 5888), or both (n = 1738) were less likely than those without prior immunity (n = 764) to report loss of taste or smell, lower respiratory tract, constitutional, or gastrointestinal symptoms and more likely to report upper respiratory tract symptoms. Stronger associations followed recent prior infection, vaccination, or more vaccine doses. Among 8539 donors without prior immunity, those with survey-reported infections during the BA.1-predominant period (n = 764) were less likely to report loss of taste or smell, or lower respiratory tract symptoms than those with infections before this period (n = 7775). Conclusions Our data suggest that both prior immunity and Omicron predominance redistributed COVID-19 symptoms toward upper respiratory tract presentations and likely both contributed to a decrease in COVID-19 severity over time. These findings may better inform COVID-19 identification in high-immunity settings and demonstrate additional benefits of vaccination.
Collapse
Affiliation(s)
- Matthew D McCullough
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia USA
| | - Bryan R Spencer
- American Red Cross, Scientific Affairs, Dedham, Massachusetts, USA
| | - Jianrong Shi
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia USA
| | - Ian D Plumb
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia USA
| | - James M Haynes
- American Red Cross, Scientific Affairs, Rockville, Maryland, USA
| | - Melisa Shah
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia USA
| | - Melissa Briggs-Hagen
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia USA
| | - Susan L Stramer
- American Red Cross, Scientific Affairs, Rockville, Maryland, USA
| | - Jefferson M Jones
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia USA
| | - Claire M Midgley
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia USA
| |
Collapse
|
17
|
Carroll M, Fox HB, Tran A, Chellappan G, Rojas LV, Karengil G, Karandish F, Langston JW, Fall BM, Whalen MM, McCluskie MJ, Durocher Y, Datta A, Kapre SV, Olave IA. SARS-CoV-2 conjugate vaccine elicits robust immune responses that can protect against evolving variants. Vaccine 2025; 54:126988. [PMID: 40054138 DOI: 10.1016/j.vaccine.2025.126988] [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: 10/08/2024] [Revised: 02/07/2025] [Accepted: 03/01/2025] [Indexed: 05/13/2025]
Abstract
The SARS-CoV-2 pandemic necessitated effective vaccines that can endure antigenic mutations. Here we demonstrate highly immunogenic conjugate vaccines that elicit broad cross-neutralization to variants of concern (VOC) in animal studies. By utilizing protein-protein conjugation and Toll-Like Receptor (TLR) agonist adjuvants we achieve enhanced immunogenicity compared to unconjugated equivalents. These vaccine candidates induced broad cross-protection against several VOC, a characteristic lacking in early COVID-19 vaccines. Murine neutralizing antibody (nAb) titers from animals vaccinated with Beta-only conjugates were equivalent between Beta, Delta, Omicron BA.1, BA.2, and BA.4/BA.5 variants, which were circulating up to three years after the antigenic Beta strain. Additionally, Beta-Delta bivalent conjugate vaccines readily prevented disease in hamster challenge. Together this demonstrates a vaccine with remarkably broad cross-protection and potential to protect for extended periods despite mutations, without requiring modified boosters or antigen adaption. These techniques can be applied to more recent SARS-CoV-2 strains, and other viruses, highlighting the benefits of protein-protein conjugation coupled with TLR agonist secondary adjuvants.
Collapse
MESH Headings
- Animals
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Mice
- Vaccines, Conjugate/immunology
- Vaccines, Conjugate/administration & dosage
- Cricetinae
- Cross Protection
- Female
- Adjuvants, Immunologic
- Spike Glycoprotein, Coronavirus/immunology
- Mice, Inbred BALB C
- Immunogenicity, Vaccine
- Humans
Collapse
Affiliation(s)
| | - Heather B Fox
- Viral Vaccines R&D, Inventprise, Inc., Redmond, WA, USA
| | - Anh Tran
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | | | | | | | | | | | - Brent M Fall
- Viral Vaccines R&D, Inventprise, Inc., Redmond, WA, USA
| | - Mary M Whalen
- Viral Vaccines R&D, Inventprise, Inc., Redmond, WA, USA
| | - Michael J McCluskie
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Yves Durocher
- Life Sciences - NRC Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC, Canada
| | - Anup Datta
- Bacterial Vaccines R&D, Inventprise, Inc., Redmond, WA, USA
| | | | - Ivan A Olave
- Viral Vaccines R&D, Inventprise, Inc., Redmond, WA, USA.
| |
Collapse
|
18
|
Tiruneh YM, Choi J, Cuccaro PM, Martinez J, Xie J, Owens M, Yamal JM. Sociodemographic and health-related predictors of COVID-19 booster uptake among fully vaccinated adults. Vaccine 2025; 54:127048. [PMID: 40157255 DOI: 10.1016/j.vaccine.2025.127048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND To control COVID-19 mutations and prevent further spread, periodic revaccination is essential. Despite the substantial evidence supporting vaccine efficacy, hesitancy towards COVID-19 booster doses persists. METHODS We examined factors associated with the booster vaccine uptake and the intention to receive a booster among fully vaccinated adults in Texas (N = 14,543), using a weighted telephone survey in 2022. We employed multiple logistic regression with Lasso-selected variables to identify sociodemographic, geographic, and health-related predictors of booster uptake and intentions. RESULTS Of the respondents, 9989 (70 %) respondents reported having received a booster dose. Booster uptake was higher among older (65 years and older), White, publicly insured, and financially stable individuals. It was also higher among those without a history of COVID-19 infection and those with comorbidities and concerns about the virus. Higher odds of being boosted were associated with older age, Asian race, Spanish language, three public health regions (PHR 11, 7, 9/10), and diabetes and HIV diagnoses. Lower odds were observed among smokers, the uninsured, and those experiencing financial struggles. Among those who had not received the booster, greater intentions to receive a booster were observed across all racial/ethnic groups compared to White individuals, Spanish speakers, and the uninsured. Those with diabetes expressed stronger intentions to receive a booster, while individuals previously infected with COVID-19 and males were less inclined to seek a booster. However, individuals expressing concerns about the infection had higher intention to be boosted. CONCLUSION The findings highlighted disparities in booster vaccinations across geographic regions, racial/ethnic groups, and insurance status. Targeted educational initiatives about the importance of booster vaccination are needed, especially for underserved populations with limited access to healthcare. Public health efforts should also focus on countering misinformation and promoting the benefits of boosters through gain-framed messages to motivate vaccine uptake and mitigate the spread of COVID-19.
Collapse
Affiliation(s)
- Yordanos M Tiruneh
- Department of Preventive Medicine and Population Health, School of Medicine, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA; Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Jihye Choi
- Center for Health Promotion and Prevention Research, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Health Promotion and Behavioral Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Paula M Cuccaro
- Center for Health Promotion and Prevention Research, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Health Promotion and Behavioral Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Journey Martinez
- Coordinating Center for Clinical Trials, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jing Xie
- Coordinating Center for Clinical Trials, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Mark Owens
- Department of Political Science, The Citadel, Charleston, SC 29409, USA
| | - Jose-Miguel Yamal
- Coordinating Center for Clinical Trials, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
19
|
Qian B, Luo R, Shen B, Fan L, Zhang J, Zhang S, Sun Y, Deng X, Pang X, Zhong W, Gao Y. EIDD-2801 resists to infection and co-infection of SARS-CoV-2 and influenza virus. Virol J 2025; 22:126. [PMID: 40296172 PMCID: PMC12039283 DOI: 10.1186/s12985-025-02755-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic has exerted a catastrophic impact on public health. Meanwhile, the seasonal influenza outbreak overlaps with the current pandemic wave. There is still an urgent need to develop effective therapeutic agents for the treatment of co-infection of multiple respiratory viruses. This study aimed to investigate antiviral effects of EIDD-2801, an orally bioavailable ribonucleoside analog, and its potent therapeutic effects in co-infection of multiple respiratory viruses. METHODS BALB/c mice and hamsters were infected with IFV or SARS-CoV-2, then were dosed orally with EIDD-2801 to measure the antiviral effects of EIDD-2801. Viral replication and mRNA transcription were evaluated by quantitative polymerase chain reaction (qPCR) and protein expression by Western Blot. Influenza viral titer was assessed using EID50 assay. RESULTS EIDD-2801 was found to be significantly effective against influenza A virus and influenza B virus. The antiviral activity against SARS-CoV-2 and further co-infection with influenza virus was also distinct. EIDD-2801 had potent antiviral effects against multiple respiratory viruses both in vitro and in vivo. CONCLUSION This study demonstrated that the small-molecule compound EIDD-2801, an orally available broad-spectrum antiviral agent, significantly inhibited the infection of influenza virus and SARS-CoV-2 and effectively protected animals from lethal influenza virus co-infection.
Collapse
Affiliation(s)
- Bingshuo Qian
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Rongbo Luo
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Beilei Shen
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Lingjun Fan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Junkui Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Shijun Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Yan Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Xiuwen Deng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Xiaobin Pang
- School of Pharmacy, Henan University, Kaifeng, 475004, China.
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Yuwei Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China.
| |
Collapse
|
20
|
Schendel SL, Yu X, Halfmann PJ, Mahita J, Ha B, Hastie KM, Li H, Bedinger D, Troup C, Li K, Kuzmina N, Torrelles JB, Munt JE, Maddocks M, Osei-Twum M, Callaway HM, Reece S, Palser A, Kellam P, Dennison SM, Huntwork RHC, Horn GQ, Abraha M, Feeney E, Martinez-Sobrido L, Pino PA, Hicks A, Ye C, Park JG, Maingot B, Periasamy S, Mallory M, Scobey T, Lepage MN, St-Amant N, Khan S, Gambiez A, Baric RS, Bukreyev A, Gagnon L, Germann T, Kawaoka Y, Tomaras GD, Peters B, Saphire EO. A global collaboration for systematic analysis of broad-ranging antibodies against the SARS-CoV-2 spike protein. Cell Rep 2025; 44:115499. [PMID: 40184253 PMCID: PMC12014896 DOI: 10.1016/j.celrep.2025.115499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/31/2025] [Accepted: 03/11/2025] [Indexed: 04/06/2025] Open
Abstract
The Coronavirus Immunotherapeutic Consortium (CoVIC) conducted side-by-side comparisons of over 400 anti-SARS-CoV-2 spike therapeutic antibody candidates contributed by large and small companies as well as academic groups on multiple continents. Nine reference labs analyzed antibody features, including in vivo protection in a mouse model of infection, spike protein affinity, high-resolution epitope binning, ACE-2 binding blockage, structures, and neutralization of pseudovirus and authentic virus infection, to build a publicly accessible dataset in the database CoVIC-DB. High-throughput, high-resolution binning of CoVIC antibodies defines a broad and predictive landscape of antibody epitopes on the SARS-CoV-2 spike protein and identifies features associated with durable potency against multiple SARS-CoV-2 variants of concern and high in vivo efficacy. Results of the CoVIC studies provide a guide for selecting effective and durable antibody therapeutics and for immunogen design as well as providing a framework for rapid response to future viral disease outbreaks.
Collapse
Affiliation(s)
- Sharon L Schendel
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Xiaoying Yu
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Peter J Halfmann
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53711, USA
| | - Jarjapu Mahita
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Brendan Ha
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Kathryn M Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Haoyang Li
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | | | | | - Kan Li
- Center for Human Systems Immunology, Departments of Surgery and Integrative Immunobiology, Duke University, Durham, NC 27701, USA
| | - Natalia Kuzmina
- Department of Pathology, University of Texas Medical Branch at Galveston, 301 University Boulevard, Galveston, TX 77555, USA; Galveston National Laboratory, 301 University Boulevard, Galveston, TX 77550, USA
| | - Jordi B Torrelles
- Disease Intervention and Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; Population Health Program, International Center for the Advancement of Research & Education (I-CARE), Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Jennifer E Munt
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| | - Melissa Maddocks
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| | - Mary Osei-Twum
- Nexelis, a Q2 Solutions Company, 525 Boulevard Cartier Ouest, Laval, QC H7V 3S8, Canada
| | - Heather M Callaway
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Stephen Reece
- Kymab, a Sanofi Company, Babraham Research Campus, Cambridge CB22 3AT, UK
| | | | - Paul Kellam
- RQ Biotechnology Ltd., London W12 7RZ, UK; Department of Infectious Diseases, Faculty of Medicine, Imperial College, London SW7 2AZ, UK
| | - S Moses Dennison
- Center for Human Systems Immunology, Departments of Surgery and Integrative Immunobiology, Duke University, Durham, NC 27701, USA
| | - Richard H C Huntwork
- Center for Human Systems Immunology, Departments of Surgery and Integrative Immunobiology, Duke University, Durham, NC 27701, USA
| | - Gillian Q Horn
- Center for Human Systems Immunology, Departments of Surgery and Integrative Immunobiology, Duke University, Durham, NC 27701, USA
| | - Milite Abraha
- Center for Human Systems Immunology, Departments of Surgery and Integrative Immunobiology, Duke University, Durham, NC 27701, USA
| | - Elizabeth Feeney
- Center for Human Systems Immunology, Departments of Surgery and Integrative Immunobiology, Duke University, Durham, NC 27701, USA
| | - Luis Martinez-Sobrido
- Disease Intervention and Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; Population Health Program, International Center for the Advancement of Research & Education (I-CARE), Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Paula A Pino
- Disease Intervention and Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Amberlee Hicks
- Disease Intervention and Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Chengjin Ye
- Disease Intervention and Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX 78227, USA; Population Health Program, International Center for the Advancement of Research & Education (I-CARE), Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Jun-Gyu Park
- Disease Intervention and Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Billie Maingot
- Disease Intervention and Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Sivakumar Periasamy
- Galveston National Laboratory, 301 University Boulevard, Galveston, TX 77550, USA
| | - Michael Mallory
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| | - Trevor Scobey
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| | - Marie-Noelle Lepage
- Nexelis, a Q2 Solutions Company, 525 Boulevard Cartier Ouest, Laval, QC H7V 3S8, Canada
| | - Natalie St-Amant
- Nexelis, a Q2 Solutions Company, 525 Boulevard Cartier Ouest, Laval, QC H7V 3S8, Canada
| | - Sarwat Khan
- Nexelis, a Q2 Solutions Company, 525 Boulevard Cartier Ouest, Laval, QC H7V 3S8, Canada
| | - Anaïs Gambiez
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Ralph S Baric
- Population Health Program, International Center for the Advancement of Research & Education (I-CARE), Texas Biomedical Research Institute, San Antonio, TX 78227, USA; Department of Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch at Galveston, 301 University Boulevard, Galveston, TX 77555, USA; Galveston National Laboratory, 301 University Boulevard, Galveston, TX 77550, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Luc Gagnon
- Nexelis, a Q2 Solutions Company, 525 Boulevard Cartier Ouest, Laval, QC H7V 3S8, Canada
| | | | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53711, USA; Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan; The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo 162-8655, Japan; Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), University of Tokyo, Tokyo 162-8655, Japan
| | - Georgia D Tomaras
- Center for Human Systems Immunology, Departments of Surgery and Integrative Immunobiology, Duke University, Durham, NC 27701, USA
| | - Bjoern Peters
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA.
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA.
| |
Collapse
|
21
|
Keeling MJ, Hill EM, Petrou S, Tran PB, Png ME, Staniszewska S, Clark C, Hassel K, Stowe J, Andrews N. Cost-effectiveness of routine COVID-19 adult vaccination programmes in England. Vaccine 2025; 53:126948. [PMID: 40023905 DOI: 10.1016/j.vaccine.2025.126948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
In England, and many other countries, immunity to SARS-CoV-2 infection and COVID-19 disease is highly heterogeneous. Immunity has been acquired through natural infection, primary and booster vaccination, while protection has been lost through waning immunity and viral mutation. During the height of the pandemic in England, the main aim was to rapidly protect the population and large supplies of vaccine were pre-purchased, eliminating the need for cost-effective calculations. As we move to an era where for the majority of the population SARS-CoV-2 infections cause relatively mild disease, and vaccine stocks need to be re-purchased, it is important we consider the cost-effectiveness and economic value of COVID-19 vaccination programmes. Here using data from 2023 and 2024 in England on COVID-19 hospital admissions, ICU admissions and deaths, coupled with bespoke health economic costs, we consider the willingness to pay threshold for COVID-19 vaccines in different age and risk groups. Willingness to pay thresholds vary from less than £1 for younger age-groups without any risk factors, to over £100 for older age-groups with comorbidities that place them at risk. This extreme non-linear dependence on age, means that despite the different method of estimating vaccine effectiveness, there is considerable qualitative agreement on the willingness to pay threshold, and therefore which ages it is cost-effective to vaccinate. The historic offer of COVID-19 vaccination to those 65 and over for the autumn 2023 programme and those over 75 for the spring 2023 programme, aligns with our cost- effective threshold for pre-purchased vaccine when the only cost was administration. However, for future programmes, when vaccine costs are included, the age-thresholds slowly increase thereby demonstrating the continued importance of protecting the eldest and most vulnerable in the population.
Collapse
Affiliation(s)
- Matt J Keeling
- The Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research, School of Life Sciences and Mathematics Institute, University of Warwick, Coventry CV4 7AL, UK.
| | - Edward M Hill
- Civic Health Innovation Labs and Institute of Population Health, University of Liverpool, Liverpool, L69 7ZX, UK; NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool, L69 7ZX, UK
| | - Stavros Petrou
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - Phuong Bich Tran
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - May Ee Png
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | | | - Corinna Clark
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Katie Hassel
- Immunisation Division, UK Health Security Agency, London, E14 4PU, UK
| | - Julia Stowe
- Immunisation Division, UK Health Security Agency, London, E14 4PU, UK
| | - Nick Andrews
- Immunisation Division, UK Health Security Agency, London, E14 4PU, UK
| |
Collapse
|
22
|
Fan X, Zhao Y, Zhang X, Li S, Wu F, Cui M, Ye Y, Duoji W, Jiang S, Yuan J, Sun C. Vaccination burnout impedes the compliance with multiple-dose administration of vaccines. Sci Rep 2025; 15:13269. [PMID: 40246920 PMCID: PMC12006531 DOI: 10.1038/s41598-025-97959-4] [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: 07/11/2024] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
Timely and complete administration of multiple-dose vaccines is essential to guarantee the efficacy. Our study aimed to investigate how people's vaccination attitudes changed over the course of the multi-dose vaccination schedule and comprehensively reported a phenomenon of vaccination burnout. The participants' vaccination burnout levels were quantified by our designed vaccination burnout scale. This study is a retrospective cross-sectional study. Among 3068 valid participants, 2991 had finished the routine two shots of COVID-19 vaccine, and 2367 had a positive attitude towards the primary doses of vaccination. Notably, 232 participants who previously had a positive attitude towards primary doses of vaccination refused to take additional multiple shots, and another 83 had changed their positive attitudes to negative, despite having taken the third shot. Participants whose attitudes or behaviors had changed had higher scores of vaccination burnout than those who still maintained a positive attitude (Z=-8.491, P < .001). The frequency of actively paying attention to the related disease news, occupation, monthly income, and residence of the participants were key factors associated with the vaccination burnout. Interventions should be implemented to alleviate the exhausted attitudes and improve people's compliance with vaccination schedules against the future pandemic.
Collapse
Affiliation(s)
- Xueying Fan
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yangguo Zhao
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Xinyu Zhang
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Shunran Li
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Fan Wu
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Mingting Cui
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Ying Ye
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Wangmu Duoji
- Tibet Center for Disease Control and Prevention, Lhasa, 850000, China
| | - Shiqiang Jiang
- Nanshan District Center for Disease Control and Prevention, Shenzhen, 518000, China
| | - Jianhui Yuan
- Nanshan District Center for Disease Control and Prevention, Shenzhen, 518000, China.
| | - Caijun Sun
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
23
|
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
|
24
|
Bao H, Meng H, Gong S, Gong Y, Tu G, Du Z, Wang Y, Wu J, Ma C, Ma Q, Yao X. Design, synthesis and activity evaluation of 4-(quinoline-2-yl)aniline derivatives as SARS-CoV‑2 main protease inhibitors. Bioorg Med Chem 2025; 121:118135. [PMID: 40024142 DOI: 10.1016/j.bmc.2025.118135] [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: 12/05/2024] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
Since 2020, numerous compounds have been investigated for their potential use in treating SARS-CoV-2 infections. By identifying the molecular targets during the virus replication process, rationally designed anti-SARS-CoV-2 agents are developed. Among these targets, the main protease (Mpro) is a crucial enzyme required for virus replication, and its highly conserved characteristic make it an important drug target for the development of anti-SARS-CoV-2 drugs. Herein, we utilized warhead-based design strategy to conduct the structural optimization of M-1 developed through virtual screening, leading to a series of novel Mpro inhibitors with 4-(quinolin-2-yl)aniline scaffold. Among them, M-32 exhibited good SARS-CoV-2 Mpro inhibitory activity (IC50 = 5.2 μM) with a nearly 25-fold increase. Isothermal titration calorimetry (ITC) directly proved that M-32 binds directly to SARS-CoV-2 Mpro in an entropy-driven manner. Mass spectrometry (MS) further confirmed the covalent binding ability of M-32 to Mpro. Meanwhile, M-32 effectively inhibited the replication of SARS-CoV-2 in Vero E6 cells (EC50 = 5.29 μM).
Collapse
Affiliation(s)
- Honglei Bao
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Hui Meng
- Collaborative Innovation Centre of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Research Centre of Chiral Hydroxyl Pharmaceutical, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Shilin Gong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Yaguo Gong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Gao Tu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Zhenya Du
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China; Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China
| | - Yuwei Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Jianlin Wu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Chunhua Ma
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China; Collaborative Innovation Centre of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Research Centre of Chiral Hydroxyl Pharmaceutical, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China.
| | - Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Xiaojun Yao
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China.
| |
Collapse
|
25
|
Xu J, Wang B, Zhao Z, Wu S, Zhang Z, Liu S, Huo N, Zheng W, Chen Y, Gao Z, Jia Z, Liu T, Zhu L, Hou L. Development of a novel adenovirus type 4 vector as a promising respiratory vaccine vehicle. Front Immunol 2025; 16:1572081. [PMID: 40276512 PMCID: PMC12018414 DOI: 10.3389/fimmu.2025.1572081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/20/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction Adenovirus (Ad) vectors are widely used for gene delivery, and some of them have been approved for vaccine development. In particular, the recombinant COVID-19 vaccine for inhalation, which was developed using adenovirus type 5 (Ad5), represents a milestone in respiratory immunization. Owing to the high pre-existing immunity (PEI) to Ad5, the development of an adenoviral vector with lower PEI and higher immunogenicity has been explored. However, the majority of the developed novel Ad vectors showed suboptimal immunogenicity compared to Ad5 in animal models. Method In this study, we constructed a novel replication-deficient viral vector based on human adenovirus type 4 (Ad4), which has long been used as a live virus vaccine with a favorable safety profile in the U.S. military. The mice were immunized intramuscularly or intranasally with an Ad4-vectored vaccine to verify immune responses and protective efficacy. Results Compared with Ad5, the novel Ad4 vector showed comparable viral growth kinetics and transgene expression in cells and similar exogenous protein expression and distribution in mice. Furthermore, the Ad4-vectored vaccine elicited superior humoral and cellular responses and protective effects when vaccinated intranasally than those triggered by the Ad5-vectored vaccine. Finally, the heterologous Ad5 prime and Ad4 boost immunization showed better immunogenicity and protective efficacy. Discussion This study broadens the research trajectory of adenovirus-vectored vaccines and offers a new option for the development of recombinant viral-vectored vaccines.
Collapse
Affiliation(s)
| | - Busen Wang
- *Correspondence: Lihua Hou, ; Li Zhu, ; Busen Wang,
| | | | | | | | | | | | | | | | | | | | | | - Li Zhu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology,
Beijing, China
| | - Lihua Hou
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology,
Beijing, China
| |
Collapse
|
26
|
Jang G, Kim J, Thompson RN, Lee H. Modeling vaccination prioritization strategies for post-pandemic COVID-19 in the Republic of Korea accounting for under-reporting and age-structure. J Infect Public Health 2025; 18:102688. [PMID: 39913986 DOI: 10.1016/j.jiph.2025.102688] [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: 11/29/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Vaccination has played a key role in limiting the impacts of COVID-19. Even though the acute phase of the COVID-19 pandemic is now over, the potential for substantial numbers of cases and deaths due to novel SARS-CoV-2 variants remains. In the Republic of Korea, a strategy of vaccinating individuals in high-risk groups annually began in October 2023. METHODS We used mathematical modeling to assess the effectiveness of alternative vaccination strategies under different assumptions about the number of available vaccine doses. An age-structured transmission model was developed using vaccination and seropositivity data. Various vaccination scenarios were considered, taking into account the effect of undetected or unreported cases (with different levels of reporting by age group): S1: prioritizing vaccination towards the oldest individuals; S2: prioritizing vaccination towards the youngest individuals; and S3: spreading vaccines among all age groups. RESULTS Our analysis reveals three key findings. First, administering vaccines to older age groups reduces the number of deaths, while instead targeting younger individuals reduces the number of infections. Second, with approximately 6,000,000 doses available annually, it is recommended that older age groups are prioritized for vaccination, achieving a substantial reduction in the number of deaths compared to a scenario without vaccination. Finally, since case detection (and subsequent isolation) affects transmission, the number of cumulative cases was found to be affected substantially by changes in the reporting rate. CONCLUSIONS In conclusion, vaccination and case detection (facilitated by contact tracing) both play important roles in limiting the impacts of COVID-19. The mathematical modeling approach presented here provides a framework for assessing the effectiveness of different vaccination strategies in scenarios with limited vaccine supply.
Collapse
Affiliation(s)
- Geunsoo Jang
- Nonlinear Dynamics and Mathematical Application Center, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jihyeon Kim
- Department of Statistics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Robin N Thompson
- Mathematical Institute, University of Oxford, Oxford OX2 6GG, United Kingdom
| | - Hyojung Lee
- Department of Statistics, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
27
|
Bendall EE, Dimcheff D, Papalambros L, Fitzsimmons WJ, Zhu Y, Schmitz J, Halasa N, Chappell J, Martin ET, Biddle JE, Smith-Jeffcoat SE, Rolfes MA, Mellis A, Talbot HK, Grijalva C, Lauring AS. In depth sequencing of a serially sampled household cohort reveals the within-host dynamics of Omicron SARS-CoV-2 and rare selection of novel spike variants. PLoS Pathog 2025; 21:e1013134. [PMID: 40294030 PMCID: PMC12074595 DOI: 10.1371/journal.ppat.1013134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 05/13/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
SARS-CoV-2 has undergone repeated and rapid evolution to circumvent host immunity. However, outside of prolonged infections in immunocompromised hosts, within-host positive selection has rarely been detected. Here we combine daily longitudinal sampling of individuals with replicate sequencing to increase the accuracy of and lower the threshold for variant calling. We sequenced 577 specimens from 105 individuals in a household cohort during the BA.1/BA.2 variant period. Individuals exhibited extremely low viral diversity, and we estimated a low within-host evolutionary rate. Within-host dynamics were dominated by genetic drift and purifying selection. Positive selection was rare but highly concentrated in spike. A Wright Fisher Approximate Bayesian Computational model identified positive selection at 14 loci with 7 in spike, including S:448 and S:339. This detectable immune-mediated selection is unusual in acute respiratory infections and may be caused by the relatively narrow antibody repertoire in individuals during the early Omicron phase of the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Emily E. Bendall
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Derek Dimcheff
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Leigh Papalambros
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - William J. Fitzsimmons
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yuwei Zhu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jonathan Schmitz
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Natasha Halasa
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - James Chappell
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Emily T. Martin
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jessica E. Biddle
- Centers for Disease Control and Prevention, Atlanta, GeorgiaUnited States of America
| | | | - Melissa A. Rolfes
- Centers for Disease Control and Prevention, Atlanta, GeorgiaUnited States of America
| | - Alexandra Mellis
- Centers for Disease Control and Prevention, Atlanta, GeorgiaUnited States of America
| | - H. Keipp Talbot
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, Tennessee Tennessee, United States of America
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Carlos Grijalva
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Adam S. Lauring
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
28
|
Iba A, Hosozawa M, Hori M, Muto Y, Kihara T, Muraki I, Masuda R, Tamiya N, Iso H. Booster vaccination and post-COVID-19 condition during the Omicron variant-dominant wave: a large population-based study. Clin Microbiol Infect 2025; 31:630-635. [PMID: 39662823 DOI: 10.1016/j.cmi.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
OBJECTIVES The effect of the COVID-19 booster vaccination and the long-term consequences concerning preventing post-COVID-19 condition (PCC) remains unclear. We aimed to investigate the association of COVID-19 booster vaccination dose and vaccination timing before infection with the risk of PCC during the Omicron variant-dominant wave. METHODS This population-based study included patients confirmed with COVID-19 (extracted from the Health Center Real-time Information-sharing System) aged 20-69 years, who were infected between 1 July and 31 August 2022. We used a self-report questionnaire to evaluate PCC and extracted information on vaccination from the municipal vaccine registry system. We calculated multiple propensity scores for COVID-19 vaccination status (unvaccinated, 1-2 doses and ≥3 doses) to control for baseline population differences. We then used a logistic regression model with inverse probability weighting to analyse the associations between the number of vaccine doses and the risk of PCC. Additionally, we conducted stratified analysis by gender and subgroup analysis for respiratory and neurological symptoms. Multivariable logistic regression was used to analyse the association between vaccination timing and PCC risk, adjusting for vaccination doses. RESULTS Of the 7936 participants with COVID-19 (mean age 42.9 years, 4553 women), 940 (11.8%) had at least 1 PCC. Compared with people unvaccinated, those vaccinated ≥3 times before the infection had a lower probability of PCC with the OR of 0.69 (95% CI: 0.53-0.90), although we detected no association with one or two doses. This association was present in women (≥3 doses vs. unvaccinated OR: 0.70, 95% CI: 0.51-0.95) but not in men. Those vaccinated ≥3 times had fewer neurological symptoms compared with those unvaccinated (OR: 0.61, 95% CI: 0.45-0.83); however, no significant association was found for respiratory symptoms. DISCUSSION This study suggests that booster vaccination could lower the risk of PCC.
Collapse
Affiliation(s)
- Arisa Iba
- Institute of Global Health Policy Research, Bureau of International Health Cooperation, National Center for Global Health and Medicine, Shinjuku, Tokyo, Japan.
| | - Mariko Hosozawa
- Institute of Global Health Policy Research, Bureau of International Health Cooperation, National Center for Global Health and Medicine, Shinjuku, Tokyo, Japan
| | - Miyuki Hori
- Institute of Global Health Policy Research, Bureau of International Health Cooperation, National Center for Global Health and Medicine, Shinjuku, Tokyo, Japan
| | - Yoko Muto
- Institute of Global Health Policy Research, Bureau of International Health Cooperation, National Center for Global Health and Medicine, Shinjuku, Tokyo, Japan
| | - Tomomi Kihara
- Institute of Global Health Policy Research, Bureau of International Health Cooperation, National Center for Global Health and Medicine, Shinjuku, Tokyo, Japan; Department of Public Health Medicine, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan; Health Services Research and Development Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Isao Muraki
- Public Health, Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Rie Masuda
- Health Services Research and Development Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Nanako Tamiya
- Health Services Research and Development Center, University of Tsukuba, Tsukuba, Ibaraki, Japan; Department of Health Services Research, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiroyasu Iso
- Institute of Global Health Policy Research, Bureau of International Health Cooperation, National Center for Global Health and Medicine, Shinjuku, Tokyo, Japan
| |
Collapse
|
29
|
Benlarbi M, Kenfack DD, Dionne K, Côté-Chenette M, Beaudoin-Bussières G, Bélanger É, Ding S, Goni OH, Ngoume YF, Tauzin A, Medjahed H, Ghedin E, Duerr R, Finzi A, Tongo M. Longitudinal humoral immunity against SARS-CoV-2 Spike following infection in individuals from Cameroon. Virology 2025; 605:110467. [PMID: 40037139 PMCID: PMC11937844 DOI: 10.1016/j.virol.2025.110467] [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: 10/30/2024] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
In May 2023 the World Health Organization (WHO) declared the end of COVID-19 as a public health emergency. Seroprevalence studies performed in African countries, such as Cameroon, depicted a much higher COVID-19 burden than reported by the WHO. To better understand humoral responses kinetics following infection, we enrolled 333 participants from Yaoundé, Cameroon between March 2020 and January 2022. We measured the levels of antibodies targeting the SARS-CoV-2 receptor-binding-domain (RBD) and the Spike glycoproteins of Delta, Omicron BA.1 and BA.4/5 and the common cold coronavirus HCoV-OC43. We also evaluated plasma capacity to neutralize authentic SARS-CoV-2 virus and to mediate Antibody-Dependent Cellular Cytotoxicity (ADCC). Most individuals mounted a strong antibody response against SARS-CoV-2 Spike. Plasma neutralization waned faster than anti-Spike binding and ADCC. We observed differences in humoral responses by age and circulating variants. Altogether, we show a global overview of antibody dynamics and functionality against SARS-CoV-2 in Cameroon.
Collapse
Affiliation(s)
- Mehdi Benlarbi
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Dell-Dylan Kenfack
- Center of Research for Emerging and Re-Emerging Diseases (CREMER), Institute of Medical Research and Study of Medicinal Plants (IMPM), Yaoundé, Cameroon
| | - Katrina Dionne
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Maxime Côté-Chenette
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Guillaume Beaudoin-Bussières
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Étienne Bélanger
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Shilei Ding
- Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Oumarou H Goni
- Center of Research for Emerging and Re-Emerging Diseases (CREMER), Institute of Medical Research and Study of Medicinal Plants (IMPM), Yaoundé, Cameroon
| | - Yannick F Ngoume
- Center of Research for Emerging and Re-Emerging Diseases (CREMER), Institute of Medical Research and Study of Medicinal Plants (IMPM), Yaoundé, Cameroon
| | - Alexandra Tauzin
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Halima Medjahed
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Elodie Ghedin
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, MD, USA
| | - Ralf Duerr
- Vaccine Center, NYU Grossman School of Medicine, New York, USA; Department of Medicine, NYU Grossman School of Medicine, New York, USA; Department of Microbiology, NYU Grossman School of Medicine, New York, USA
| | - Andrés Finzi
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada.
| | - Marcel Tongo
- Center of Research for Emerging and Re-Emerging Diseases (CREMER), Institute of Medical Research and Study of Medicinal Plants (IMPM), Yaoundé, Cameroon; HIV Pathogenesis Program, The Doris Duke Medical Research Institute, University of KwaZulu Natal, Durban, South Africa.
| |
Collapse
|
30
|
Ye ZW, Ong CP, Cao H, Tang K, Gray VS, Hinson Cheung PH, Wang J, Li W, Zhang H, Luo P, Ni T, Chan CP, Zhang M, Zhang Y, Ling GS, Yuan S, Jin DY. A live attenuated SARS-CoV-2 vaccine constructed by dual inactivation of NSP16 and ORF3a. EBioMedicine 2025; 114:105662. [PMID: 40132472 PMCID: PMC11985078 DOI: 10.1016/j.ebiom.2025.105662] [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: 06/19/2024] [Revised: 02/16/2025] [Accepted: 03/08/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Live attenuated vaccines against SARS-CoV-2 activate all phases of host immunity resembling a natural infection and they block viral transmission more efficiently than existing vaccines in human use. In our prior work, we characterised an attenuated SARS-CoV-2 variant, designated d16, which harbours a D130A mutation in the NSP16 protein, inactivating its 2'-O-methyltransferase function. The d16 variant has demonstrated an ability to induce both mucosal and sterilising immunity in animal models. However, further investigation is required to identify any additional modifications to d16 that could mitigate concerns regarding potential virulence reversion and the suboptimal regulation of the proinflammatory response. METHODS Mutations were introduced into molecular clone of SARS-CoV-2 and live attenuated virus was recovered from cultured cells. Virological, biochemical and immunological assays were performed in vitro and in two animal models to access the protective efficacies of the candidate vaccine strain. FINDINGS Here we describe evaluation of a derivative of d16. We further modified the d16 variant by inverting the open reading frame of the ORF3a accessory protein, resulting in the d16i3a strain. This modification is anticipated to enhance safety and reduce pathogenicity. d16i3a appeared to be further attenuated in hamsters and transgenic mice compared to d16. Intranasal vaccination with d16i3a stimulated humoural, cell-mediated and mucosal immune responses, conferring sterilising protection against SARS-CoV-2 Delta and Omicron variants in animals. A version of d16i3a expressing the XBB.1.16 spike protein further expanded the vaccine's protection spectrum against circulating variants. Notably, this version has demonstrated efficacy as a booster in hamsters, providing protection against Omicron subvariants and achieving inhibition of viral transmission. INTERPRETATION Our work established a platform for generating safe and effective live attenuated vaccines by dual inactivation of NSP16 and ORF3a of SARS-CoV-2. FUNDING This work was supported by National Key Research and Development Program of China (2021YFC0866100, 2023YFC3041600, and 2023YFE0203400), Hong Kong Health and Medical Research Fund (COVID190114, CID-HKU1-9, and 23220712), Hong Kong Research Grants Council (C7142-20GF and T11-709/21-N), Hong Kong Innovation and Technology Commission grant (MHP/128/22), Guangzhou Laboratory (EKPG22-01) and Health@InnoHK (CVVT). Funding sources had no role in the writing of the manuscript or the decision to submit it for publication.
Collapse
Affiliation(s)
- Zi-Wei Ye
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Chon Phin Ong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Hehe Cao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 102 Pokfulam Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Kaiming Tang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 102 Pokfulam Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Victor Sebastien Gray
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Pak-Hin Hinson Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Junjue Wang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Weixin Li
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Hongzhuo Zhang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Peng Luo
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 102 Pokfulam Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Tao Ni
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Chi Ping Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Ming Zhang
- State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, China National Biotec Group Company Limited, Beijing, 100024, China
| | - Yuntao Zhang
- State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, China National Biotec Group Company Limited, Beijing, 100024, China
| | - Guang Sheng Ling
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Shuofeng Yuan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 102 Pokfulam Road, Pokfulam, Hong Kong Special Administrative Region of China
| | - Dong-Yan Jin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region of China.
| |
Collapse
|
31
|
Uehara T, Yotsuyanagi H, Ohmagari N, Doi Y, Yamato M, Imamura T, Sakaguchi H, Fukushi A, Takeda Y, Baba K, Nobori H, Miyamoto T, Arita S, Dodo R, Shimba A, Fukao K, Shishido T, Tsuge Y, Mukae H. Ensitrelvir treatment-emergent amino acid substitutions in SARS-CoV-2 3CL pro detected in the SCORPIO-SR phase 3 trial. Antiviral Res 2025; 236:106097. [PMID: 39892563 DOI: 10.1016/j.antiviral.2025.106097] [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: 11/11/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
The impact of treatment-emergent amino acid substitutions (TEAASs) in severe acute respiratory syndome coronavirus 2 (SARS-CoV-2) 3C-like protease (3CLpro) on clinical and virologic outcomes was evaluated in patients with mild-to-moderate coronavirus disease 2019 (COVID-19) who received ensitrelvir 125 mg in the SCORPIO-SR trial. Individuals were randomised to ensitrelvir or matched placebo once daily for 5 days (first dose <72 h after disease onset). 3CLpro-TEAASs were identified by sequencing nsp5 encoding 3CLpro from pre- and post-treatment nasopharyngeal swabs. Time to resolution of a composite of five characteristic COVID-19 symptoms (TTR) was compared between patients with and without the most common 3CLpro-TEAASs in the ensitrelvir arm. The ensitrelvir and placebo intention-to-treat populations comprised 345 and 341 patients, respectively. 3CLpro-TEAASs were detected in 19/204 (9.3%) ensitrelvir-treated and 3/137 (2.2%) placebo-treated patients with paired sequence data. The most common 3CLpro-TEAASs in the ensitrelvir arm were M49L (n = 12), M49I (n = 3) and S144A (n = 2). In the placebo arm, all 3CLpro-TEAASs occurred in ≤1 patient. Median (95% confidence interval) TTR was comparable between patients with and without those TEAASs (158.8 h [112.1-281.9] vs 189.7 h [151.4-234.4]). Mean viral RNA levels declined more slowly in patients with M49L/I or S144A versus those without. Reductions in viral titre were unaffected by these TEAASs. The characteristics of recombinant SARS-CoV-2 with 3CLpro mutations were explored in vitro. Recombinant viruses with some 3CLpro mutations had reduced susceptibility to ensitrelvir in vitro, with limited effects on viral and competitive fitness. Continued surveillance is warranted to monitor the spread of viruses with 3CLpro mutations.
Collapse
Affiliation(s)
- Takeki Uehara
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan.
| | | | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health, Tokyo, Japan
| | - Yohei Doi
- Departments of Microbiology and Infectious Diseases, Fujita Health University School of Medicine, Toyoake, Japan
| | - Masaya Yamato
- Department of General Medicine and Infectious Diseases, Rinku General Medical Center, Izumisano, Japan
| | - Takumi Imamura
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Hiroki Sakaguchi
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Akimasa Fukushi
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Yosuke Takeda
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Keiko Baba
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Haruaki Nobori
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Tadashi Miyamoto
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Shuhei Arita
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Reiko Dodo
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Alice Shimba
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Keita Fukao
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Takao Shishido
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Yuko Tsuge
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
32
|
Teoh YC, Noor MS, Aghakhani S, Girton J, Hu G, Chowdhury R. Viral escape-inspired framework for structure-guided dual bait protein biosensor design. PLoS Comput Biol 2025; 21:e1012964. [PMID: 40233103 PMCID: PMC12021294 DOI: 10.1371/journal.pcbi.1012964] [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: 09/03/2024] [Revised: 04/24/2025] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
A generalizable computational platform, CTRL-V (Computational TRacking of Likely Variants), is introduced to design selective binding (dual bait) biosensor proteins. The iteratively evolving receptor binding domain (RBD) of SARS-CoV-2 spike protein has been construed as a model dual bait biosensor which has iteratively evolved to distinguish and selectively bind to human entry receptors and avoid binding neutralizing antibodies. Spike RBD prioritizes mutations that reduce antibody binding while enhancing/ retaining binding with the ACE2 receptor. CTRL-V's through iterative design cycles was shown to pinpoint 20% (of the 39) reported SARS-CoV-2 point mutations across 30 circulating, infective strains as responsible for immune escape from commercial antibody LY-CoV1404. CTRL-V successfully identifies ~70% (five out of seven) single point mutations (371F, 373P, 440K, 445H, 456L) in the latest circulating KP.2 variant and offers detailed structural insights to the escape mechanism. While other data-driven viral escape variant predictor tools have shown promise in predicting potential future viral variants, they require massive amounts of data to bypass the need for physics of explicit biochemical interactions. Consequently, they cannot be generalized for other protein design applications. The publicly availably viral escape data was leveraged as in vivo anchors to streamline a computational workflow that can be generalized for dual bait biosensor design tasks as exemplified by identifying key mutational loci in Raf kinase that enables it to selectively bind Ras and Rap1a GTP. We demonstrate three versions of CTRL-V which use a combination of integer optimization, stochastic sampling by PyRosetta, and deep learning-based ProteinMPNN for structure-guided biosensor design.
Collapse
Affiliation(s)
- Yee Chuen Teoh
- Department of Computer Science, Iowa State University, Ames, Iowa, United States of America
| | - Mohammed Sakib Noor
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, United States of America
| | - Sina Aghakhani
- School of Industrial Engineering and Management, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Jack Girton
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, United States of America
| | - Guiping Hu
- School of Industrial Engineering and Management, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Ratul Chowdhury
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, United States of America
- Nanovaccine Institute, Iowa State University, Ames, Iowa, United States of America
| |
Collapse
|
33
|
Chen K, Enns EA. Evaluating trade-offs between COVID-19 prevention and learning loss: an agent-based simulation analysis. ROYAL SOCIETY OPEN SCIENCE 2025; 12:231842. [PMID: 40271137 PMCID: PMC12015571 DOI: 10.1098/rsos.231842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/07/2025] [Accepted: 03/05/2025] [Indexed: 04/25/2025]
Abstract
The COVID-19 pandemic presented significant challenges in educational settings. Schools implemented a variety of COVID-19 mitigation strategies, some of which were controversial due to potential disruptions to in-person learning. We developed an agent-based model of COVID-19 in a US high school setting to evaluate potential trade-offs between preventing COVID-19 infections versus avoiding in-person learning loss under different mitigation policies in a post-Omicron context. Mitigation policies included isolation alone and in combination with quarantine of exposed students, weekly testing of all students or testing of exposed students ('test-to-stay') under different scenarios of mask use and booster dose uptake. Outcomes were simulated over an 11 week trimester. We found that requiring a full 5 or 10 day quarantine of exposed students reduced COVID-19 infections by five to sevenfold relative to isolation alone, but at a cost of nearly 40% learning days lost. Test-to-stay achieved nearly the same level of infection reduction with lower levels of learning loss. Weekly testing also reduced COVID-19 infections but was less effective and incurred higher learning loss than test-to-stay. Universal masking and increased vaccination not only reduced infections at no cost to learning but also synergized with other strategies to reduce trade-offs.
Collapse
Affiliation(s)
| | - Eva A. Enns
- Division of Health Policy and Management, University of Minnesota School of Public Health, Minneapolis, MN, USA
| |
Collapse
|
34
|
Case JB, Jain S, Suthar MS, Diamond MS. SARS-CoV-2: The Interplay Between Evolution and Host Immunity. Annu Rev Immunol 2025; 43:29-55. [PMID: 39705164 DOI: 10.1146/annurev-immunol-083122-043054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
The persistence of SARS-CoV-2 infections at a global level reflects the repeated emergence of variant strains encoding unique constellations of mutations. These variants have been generated principally because of a dynamic host immune landscape, the countermeasures deployed to combat disease, and selection for enhanced infection of the upper airway and respiratory transmission. The resulting viral diversity creates a challenge for vaccination efforts to maintain efficacy, especially regarding humoral aspects of protection. Here, we review our understanding of how SARS-CoV-2 has evolved during the pandemic, the immune mechanisms that confer protection, and the impact viral evolution has had on transmissibility and adaptive immunity elicited by natural infection and/or vaccination. Evidence suggests that SARS-CoV-2 evolution initially selected variants with increased transmissibility but currently is driven by immune escape. The virus likely will continue to drift to maintain fitness until countermeasures capable of disrupting transmission cycles become widely available.
Collapse
Affiliation(s)
- James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| | - Shilpi Jain
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael S Diamond
- Department of Pathology & Immunology; Department of Molecular Microbiology; and Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| |
Collapse
|
35
|
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
|
36
|
Schmidt KG, Geißler P, Schuster EM, Schülein C, Harrer EG, Schönau V, Luber M, Spriewald B, Steininger P, Bergmann S, Ensser A, Schober K, Nganou-Makamdop K, Harrer T. Coronavirus replicase epitopes induce cross-reactive CD8 T cell responses in SARS-CoV-2-naive people with HIV-1. iScience 2025; 28:111949. [PMID: 40034846 PMCID: PMC11872457 DOI: 10.1016/j.isci.2025.111949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/12/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
Cross-reactive T cell immunity between common cold coronaviruses and SARS-CoV-2 may influence COVID-19 susceptibility. To identify cross-reactive CD8 T cell epitopes, we analyzed responses to 21 homologous SARS-CoV-2 replicase peptides in 177 people living with HIV (PLWH) on antiretroviral therapy, of which 133 did not have prior SARS-CoV-2 infection. Replicase peptides induced IFN-γ responses in 63% of the SARS-CoV-2-naïve individuals and in 73% of individuals with prior SARS-CoV-2-infection. We could define several cross-reactive epitopes, including the HLA-B∗35:03 restricted CoV-YL8, and characterized a CoV-YL8-specific T cell receptor cloned from a SARS-CoV-2 seronegative individual. Analysis of the association between HLA-I alleles and SARS-CoV-2 infections over a 16-months period revealed that in a cohort of 452 PLWH, HLA-B∗35:03 and C∗07 were underrepresented in the 55 persons with a history of SARS-CoV-2 infection while HLA-B∗35:01 and HLA-C∗04 were associated with a higher infection rate. Taken together, our study suggests an HLA-I-mediated effect of common cold coronaviruses on SARS-CoV-2 immunity.
Collapse
Affiliation(s)
- Katja G. Schmidt
- Infectious Diseases and Immunodeficiency Section, Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Paulina Geißler
- Infectious Diseases and Immunodeficiency Section, Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Ev-Marie Schuster
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Christine Schülein
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Ellen G. Harrer
- Infectious Diseases and Immunodeficiency Section, Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Verena Schönau
- Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Markus Luber
- Department of Medicine 5, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Bernd Spriewald
- Department of Medicine 5, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- FAU Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Steininger
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Silke Bergmann
- Infectious Diseases and Immunodeficiency Section, Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Armin Ensser
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Kilian Schober
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- FAU Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Krystelle Nganou-Makamdop
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Thomas Harrer
- Infectious Diseases and Immunodeficiency Section, Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- FAU Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
37
|
Aso S, Ono S, Michihata N, Uemura K, Yasunaga H. Differences in Characteristics, Treatments, and Mortality of Patients with COVID-19 Between 2022 and 2020-2021. Jpn J Infect Dis 2025; 78:85-90. [PMID: 39617481 DOI: 10.7883/yoken.jjid.2024.272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
In 2021, vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) were developed and the Omicron variant emerged. This study compared the characteristics, treatments, and mortality of patients with coronavirus disease 2019 (COVID-19) between 2022 and 2020-2021, using administrative claims data linked including vaccine records in a Japanese city. Patients who underwent COVID-19 antigen or polymerase chain reaction tests and were diagnosed with COVID-19 were identified. Patient characteristics, treatments, and mortality were compared between 2022 and 2020-2021 among those diagnosed with COVID-19. We identified 26,262 patients with COVID-19. The mortality in 2022 was lower than that in 2020-2021 (0.6% vs. 1.7%; P < 0.01). Patients in 2022 were significantly less likely to receive oxygen therapy, high-flow nasal oxygenation, mechanical ventilation, steroids, and tocilizumab than those in 2020-2021. Among the deceased, the proportion of those aged ≥65 years was significantly higher in 2022 than in 2020-2021 (98.4% vs. 88.6%). The logistic regression analysis indicated, older age, male sex, and ≥3 comorbidities were associated with higher mortality, whereas ≥3 vaccinations were associated with lower mortality. Patients with COVID-19 in 2022 were less likely to require respiratory care or succumb to the disease. Older patients were more likely to die in 2022 than in 2020-2021.
Collapse
Affiliation(s)
- Shotaro Aso
- Department of Health Services Research, School of Public Health, The University of Tokyo, Japan
| | - Sachiko Ono
- Department of Eat-loss Medicine, Graduate School of Medicine, School of Public Health, The University of Tokyo, Japan
| | - Nobuaki Michihata
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Japan
| | - Kohei Uemura
- Department of Biostatistics and Bioinformatics, Interfaculty Initiative in Information Studies, School of Public Health, The University of Tokyo, Japan
| | - Hideo Yasunaga
- Department of Clinical Epidemiology and Health Economics, School of Public Health, The University of Tokyo, Japan
| |
Collapse
|
38
|
Piano Mortari E, Ferrucci F, Zografaki I, Carsetti R, Pacelli L. T and B cell responses in different immunization scenarios for COVID-19: a narrative review. Front Immunol 2025; 16:1535014. [PMID: 40170841 PMCID: PMC11959168 DOI: 10.3389/fimmu.2025.1535014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/25/2025] [Indexed: 04/03/2025] Open
Abstract
Vaccines against COVID-19 have high efficacy and low rates of adverse events. However, none of the available vaccines provide sterilizing immunity, and reinfections remain possible. This review aims to summarize the immunological responses elicited by different immunization strategies, examining the roles of homologous and heterologous vaccination and hybrid immunity. Homologous vaccination regimens exhibit considerable variation in immune responses depending on the vaccine platform, particularly concerning antibody titers, B cell activation, and T cell responses. mRNA vaccines, such as mRNA-1273 and BNT162b2, consistently generate higher and more durable levels of neutralizing antibodies and memory B cells compared to adenovirus-based vaccines like Ad26.COV2.S and ChAdOx1. The combination of two distinct vaccine platforms, each targeting different immune pathways, seems to be more effective in promoting long-lasting B cell responses and potent T cell responses. The high heterogeneity of the available studies, the different dosing schemes, the succession of new variants, and the subjects' immunological background do not allow for a definitive conclusion. Overall, heterologous vaccination strategies, combining sequentially viral vector and mRNA may deliver a more balanced and robust humoral and cellular immune response compared to homologous regimens. Hybrid immunity, which arises from SARS-CoV-2 infection preceded or followed by vaccination produces markedly stronger immune responses than either vaccination or infection alone. The immune response to SARS-CoV-2 variants of concern varies depending on both the vaccine platform and prior infection status. Hybrid immunity leads to a broader antibody repertoire, providing enhanced neutralization of variants of concern. Heterologous vaccination and hybrid immunity may provide further opportunities to enhance immune responses, offering broader protection and greater durability of immunity. However, from all-cause mortality, symptomatic or severe COVID, and serious adverse events at present it is not possible to infer different effects between homologous and heterologous schemes. Next-generation vaccines could involve tweaks to these designs or changes to delivery mechanisms that might improve performance.
Collapse
Affiliation(s)
- Eva Piano Mortari
- B Lymphocytes Unit, Bambino Gesù Children’s Hospital, istituto di ricovero e cura a carattere scientifico (IRCCS), Rome, Italy
| | | | - Irini Zografaki
- mRNA & Antivirals Medical & Scientific Affairs International Developed Markets, Pfizer, Athens, Greece
| | - Rita Carsetti
- B Lymphocytes Unit, Bambino Gesù Children’s Hospital, istituto di ricovero e cura a carattere scientifico (IRCCS), Rome, Italy
| | - Luciano Pacelli
- Medical Department, Internal Medicine, Pfizer s.r.l., Rome, Italy
| |
Collapse
|
39
|
Ejima K, Ajelli M, Singh A, Chua HK, Ponce L, Wang Y, Jeong YD, Iwami S, Shibuya K, Taniguchi K, Ohmagari N, Chia PY, Ong SWX, Tan KB, Lye DC, Young BE. Age- and vaccination status-dependent isolation guidelines based on simulation of SARS-CoV-2 Delta cases in Singapore. COMMUNICATIONS MEDICINE 2025; 5:76. [PMID: 40082681 PMCID: PMC11906760 DOI: 10.1038/s43856-025-00797-8] [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: 02/09/2024] [Accepted: 03/05/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND In the absence of effective pharmaceutical interventions early in an infectious disease outbreak, non-pharmaceutical measures, especially isolating infected individuals, critically limit its impact. The ongoing COVID-19 pandemic has sparked debates on optimal isolation guidelines. This study proposes a variable isolation period approach (variable-period approach), tailoring isolation durations for distinct population groups with varied viral load dynamics. METHODS To compare our variable-period approach with a fixed-period strategy, we developed a simulation model generating synthetic longitudinal SARS-CoV-2 viral load data. The data was generated from the viral dynamics model parameterized using SARS-CoV-2 Delta patient data in Singapore, accounting for age and vaccination status. RESULTS Findings show that age and vaccination status significantly influence viral dynamics, with younger age and vaccination linked to shorter viral shedding durations. The variable-period framework suggests longer isolation lengths for older and unvaccinated individuals. By setting the leaking risk (risk of remaining infectious at the end of isolation) below 10%, the optimal fixed-period isolation is 14 days, with an average excess isolation burden of 7.4 unnecessary days. In contrast, the variable-period guideline reduces the excess isolation burden to 6.0 days, with the optimal isolation periods ranging from 9 to 16 days, depending on the population group. We confirmed similar results when we used the effective reproduction number as an alternative to the leaking risk. CONCLUSIONS In this case, study using the SARS-CoV-2 Delta variant, our analysis demonstrates that unnecessary time spent in isolation can be reduced by adopting variable-period guidelines based on patient characteristics.
Collapse
Affiliation(s)
- Keisuke Ejima
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
- The Tokyo Foundation for Policy Research, Tokyo, Japan.
| | - Marco Ajelli
- Laboratory for Computational Epidemiology and Public Health, Department of Epidemiology and Biostatistics, Indiana University School of Public Health, Bloomington, IN, USA
| | - Ananya Singh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Hoong Kai Chua
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Luis Ponce
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Yuqian Wang
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Yong Dam Jeong
- Interdisciplinary Biology Laboratory (iBLab), Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Shingo Iwami
- Interdisciplinary Biology Laboratory (iBLab), Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
- Institute of Mathematics for Industry, Kyushu University, Fukuoka, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- NEXT-Ganken Program, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
- Interdisciplinary Theoretical and Mathematical Sciences Program (iTHEMS), RIKEN, Saitama, Japan
- Science Groove Inc, Fukuoka, Japan
| | - Kenji Shibuya
- The Tokyo Foundation for Policy Research, Tokyo, Japan
| | | | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine Hospital, Tokyo, Japan
| | - Po Ying Chia
- National Centre for Infectious Diseases, Singapore, Singapore
- Tan Tock Seng Hospital, Singapore, Singapore
| | - Sean W X Ong
- National Centre for Infectious Diseases, Singapore, Singapore
- Tan Tock Seng Hospital, Singapore, Singapore
| | - Kelvin Bryan Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Division of Communicable Disease, Ministry of Health, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - David Chien Lye
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Tan Tock Seng Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Barnaby E Young
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
- National Centre for Infectious Diseases, Singapore, Singapore.
- Tan Tock Seng Hospital, Singapore, Singapore.
| |
Collapse
|
40
|
Fujitani M, Lu X, Shinnakasu R, Inoue T, Kidani Y, Seki NM, Ishida S, Mitsuki S, Ishihara T, Aoki M, Suzuki A, Takahashi K, Takayama M, Ota T, Iwata S, Shibata RY, Sonoyama T, Ariyasu M, Kitano A, Terooatea T, Kelly Villa J, Yamashita K, Yamasaki S, Kurosaki T, Omoto S. Longitudinal analysis of immune responses to SARS-CoV-2 recombinant vaccine S-268019-b in phase 1/2 prime-boost study. Front Immunol 2025; 16:1550279. [PMID: 40109335 PMCID: PMC11919840 DOI: 10.3389/fimmu.2025.1550279] [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: 12/23/2024] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
Background The durability of vaccine-induced immune memory to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is crucial for preventing infection, especially severe disease. Methods This follow-up report from a phase 1/2 study of S-268019-b (a recombinant spike protein vaccine) after homologous booster vaccination confirms its long-term safety, tolerability, and immunogenicity. Results Booster vaccination with S-268019-b resulted in an enhancement of serum neutralizing antibody (NAb) titers and a broad range of viral neutralization. Single-cell immune profiling revealed persistent and mature antigen-specific memory B cells and T follicular helper cells, with increased B-cell receptor diversity. The expansion of B- and T-cell repertoires and presence of cross-reactive NAbs targeting conserved epitopes within the receptor-binding domain following a booster accounted for the broad-spectrum neutralizing activity. Conclusion These findings highlight the potential of S-268019-b to provide broad and robust protection against a range of SARS-CoV-2 variants, addressing a critical challenge in the ongoing fight against coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Masaya Fujitani
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Xiuyuan Lu
- Laboratory of Molecular Immunology, World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Ryo Shinnakasu
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Takeshi Inoue
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yujiro Kidani
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Naomi M. Seki
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Satoru Ishida
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Shungo Mitsuki
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | | | - Miwa Aoki
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Akio Suzuki
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Koji Takahashi
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Masahiro Takayama
- Pharmaceutical Technology Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Takeshi Ota
- Pharmaceutical Technology Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Satoshi Iwata
- Department of Microbiology, Tokyo Medical University, Tokyo, Japan
| | - Risa Yokokawa Shibata
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Takuhiro Sonoyama
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Mari Ariyasu
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | | | | | | | | | - Sho Yamasaki
- Laboratory of Molecular Immunology, World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| | - Shinya Omoto
- Vaccine Business Division, Shionogi & Co., Ltd., Osaka, Japan
| |
Collapse
|
41
|
Angeli L, Caetano CP, Franco N, Coletti P, Faes C, Molenberghs G, Beutels P, Abrams S, Willem L, Hens N. Assessing the role of children in the COVID-19 pandemic in Belgium using perturbation analysis. Nat Commun 2025; 16:2230. [PMID: 40044649 PMCID: PMC11882900 DOI: 10.1038/s41467-025-57087-z] [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: 04/25/2024] [Accepted: 02/10/2025] [Indexed: 03/09/2025] Open
Abstract
Understanding the evolving role of different age groups in virus transmission is essential for effective pandemic management. We investigated SARS-CoV-2 transmission in Belgium from November 2020 to February 2022, focusing on age-specific patterns. Using a next generation matrix approach integrating social contact data and simulating population susceptibility evolution, we performed a longitudinal perturbation analysis of the effective reproduction number to unravel age-specific transmission dynamics. From November to December 2020, adults in the [18, 60) age group were the main transmission drivers, while children contributed marginally. This pattern shifted between January and March 2021, when in-person education resumed, and the Alpha variant emerged: children aged under 12 years old were crucial in transmission. Stringent social distancing measures in March 2021 helped diminish the noticeable contribution of the [18, 30) age group. By June 2021, as the Delta variant became the predominant strain, adults aged [18, 40) years emerged as main contributors to transmission, with a resurgence in children's contribution during September-October 2021. This study highlights the effectiveness of our methodology in identifying age-specific transmission patterns.
Collapse
Affiliation(s)
- Leonardo Angeli
- Data Science Institute, I-BioStat, Hasselt University, Hasselt, Belgium.
| | - Constantino Pereira Caetano
- Center for Computational and Stochastic Mathematics, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | - Nicolas Franco
- Data Science Institute, I-BioStat, Hasselt University, Hasselt, Belgium
- Namur Institute for Complex Systems (naXys) and Department of Mathematics, University of Namur, Namur, Belgium
| | - Pietro Coletti
- Data Science Institute, I-BioStat, Hasselt University, Hasselt, Belgium
- Institute of Health and Society (IRSS), UCLouvain (Université catholique de Louvain), Brussels, Belgium
| | - Christel Faes
- Data Science Institute, I-BioStat, Hasselt University, Hasselt, Belgium
| | - Geert Molenberghs
- Data Science Institute, I-BioStat, Hasselt University, Hasselt, Belgium
- L-BioStat, KU Leuven, Leuven, Belgium
| | - Philippe Beutels
- Centre for Health Economics Research and Modelling Infectious Diseases, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Steven Abrams
- Data Science Institute, I-BioStat, Hasselt University, Hasselt, Belgium
- Department of Family Medicine and Population Health (FAMPOP), University of Antwerp, Antwerp, Belgium
| | - Lander Willem
- Centre for Health Economics Research and Modelling Infectious Diseases, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Department of Family Medicine and Population Health (FAMPOP), University of Antwerp, Antwerp, Belgium
| | - Niel Hens
- Data Science Institute, I-BioStat, Hasselt University, Hasselt, Belgium
- Centre for Health Economics Research and Modelling Infectious Diseases, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
42
|
Wu Q, Wu H, Hu Y, Zheng X, Chang F, Liu Y, Pan Z, Wang Q, Tang F, Qian J, Li Y, Huang B, Chen K, Xu J, Wang Y, Xie X, Zhao P, Wu X, Qu X, Li YP. Immune evasion of Omicron variants JN.1, KP.2, and KP.3 to the polyclonal and monoclonal antibodies from COVID-19 convalescents and vaccine recipients. Antiviral Res 2025; 235:106092. [PMID: 39864525 DOI: 10.1016/j.antiviral.2025.106092] [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: 10/14/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
The Omicron BA.2.86 subvariants, JN.1, KP.2, and KP.3, have become predominant globally, raising concerns about their immune evasion from vaccines and monoclonal antibody (mAb) treatments. These variants harbor more receptor-binding domain (RBD) mutations than the XBB and EG.5 sub-lineages, which are already known to compromise vaccine and therapeutic efficacy. We evaluated sera from individuals vaccinated with inactivated vaccines, with or without breakthrough infections, as well as COVID-19 convalescents. Our results showed a substantial decrease in serum neutralizing activity against the JN.1, KP.2, XBB.1.5, and EG.5.1 variants compared to BA.2. Additionally, we developed 19 neutralizing antibodies from memory B cells, with some retaining efficacy against earlier Omicron variants. However, potency was notably diminished against newer subvariants like BF.7, BQ.1, XBB.1.5, and BA.2.86. Of mAbs, those isolated from COVID-19 convalescents, particularly SA-3, exhibited exceptional potency across ten variants from BA.2 to KP.2, with IC50 values ranging from 0.006 to 2.546 μg/mL. However, SA-3 had lost neutralizing activity against the KP.3 due to the Q493E mutation, but the KP.3 became susceptible to neutralization by the other mAb, SA-6. In contrast, SA-6 was unable to neutralize KP.2 because of the presence of R346T mutation. Our findings underscore the importance of continuous surveillance of viral evolution and the need for updated vaccines and therapeutics to combat the ongoing evolution of SARS-CoV-2, particularly in the context of emerging variants that escape both vaccine-induced immunity and monoclonal antibody treatments.
Collapse
Affiliation(s)
- Qian Wu
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Hairuo Wu
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Yabin Hu
- College of Basic Medical Sciences, Hengyang Medical School, University of South China & MOE Key Lab of Rare Pediatric Diseases, Hengyang 421001, China; Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou 423000, China
| | - Xingyu Zheng
- College of Basic Medical Sciences, Hengyang Medical School, University of South China & MOE Key Lab of Rare Pediatric Diseases, Hengyang 421001, China
| | - Fangfang Chang
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Yongchen Liu
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhendong Pan
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
| | - Qijie Wang
- The Central Hospital of Shaoyang, Shaoyang 422099, China; Xinning Country People's Hospital, Shaoyang 422099, China
| | - Fei Tang
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Jun Qian
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuezhou Li
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Bin Huang
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Keqiu Chen
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Juan Xu
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - You Wang
- College of Basic Medical Sciences, Hengyang Medical School, University of South China & MOE Key Lab of Rare Pediatric Diseases, Hengyang 421001, China
| | - Xiangping Xie
- The Central Hospital of Shaoyang, Shaoyang 422099, China
| | - Ping Zhao
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
| | - Xu Wu
- Pulmonary and Critical Care Medicine, Hengyang Medical School, University of South China, No. 30, Jiefang Road, Shigu District, Hengyang 421000, China.
| | - Xiaowang Qu
- College of Basic Medical Sciences, Hengyang Medical School, University of South China & MOE Key Lab of Rare Pediatric Diseases, Hengyang 421001, China.
| | - Yi-Ping Li
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
43
|
Chemaitelly H, Ayoub HH, Coyle P, Tang P, Hasan MR, Yassine HM, Al Thani AA, Al-Kanaani Z, Al-Kuwari E, Jeremijenko A, Kaleeckal AH, Latif AN, Shaik RM, Abdul-Rahim HF, Nasrallah GK, Al-Kuwari MG, Butt AA, Al-Romaihi HE, Al-Thani MH, Al-Khal A, Bertollini R, Abu-Raddad LJ. Differential protection against SARS-CoV-2 reinfection pre- and post-Omicron. Nature 2025; 639:1024-1031. [PMID: 39910292 PMCID: PMC11946897 DOI: 10.1038/s41586-024-08511-9] [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: 03/07/2024] [Accepted: 10/16/2024] [Indexed: 02/07/2025]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly evolved over short timescales, leading to the emergence of more transmissible variants such as Alpha and Delta1-3. The arrival of the Omicron variant marked a major shift, introducing numerous extra mutations in the spike gene compared with earlier variants1,2. These evolutionary changes have raised concerns regarding their potential impact on immune evasion, disease severity and the effectiveness of vaccines and treatments1,3. In this epidemiological study, we identified two distinct patterns in the protective effect of natural infection against reinfection in the Omicron versus pre-Omicron eras. Before Omicron, natural infection provided strong and durable protection against reinfection, with minimal waning over time. However, during the Omicron era, protection was robust only for those recently infected, declining rapidly over time and diminishing within a year. These results demonstrate that SARS-CoV-2 immune protection is shaped by a dynamic interaction between host immunity and viral evolution, leading to contrasting reinfection patterns before and after Omicron's first wave. This shift in patterns suggests a change in evolutionary pressures, with intrinsic transmissibility driving adaptation pre-Omicron and immune escape becoming dominant post-Omicron, underscoring the need for periodic vaccine updates to sustain immunity.
Collapse
Affiliation(s)
- Hiam Chemaitelly
- Infectious Disease Epidemiology Group, Weill Cornell Medicine-Qatar, Cornell University, Qatar Foundation - Education City, Doha, Qatar.
- World Health Organization Collaborating Centre for Disease Epidemiology Analytics on HIV/AIDS, Sexually Transmitted Infections, and Viral Hepatitis, Weill Cornell Medicine-Qatar, Cornell University, Qatar Foundation - Education City, Doha, Qatar.
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| | - Houssein H Ayoub
- Mathematics Program, Department of Mathematics, Statistics and Physics, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Peter Coyle
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
- Hamad Medical Corporation, Doha, Qatar
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University, Belfast, UK
| | - Patrick Tang
- Department of Pathology, Sidra Medicine, Doha, Qatar
| | - Mohammad R Hasan
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Hadi M Yassine
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
| | - Asmaa A Al Thani
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
| | | | | | | | | | | | | | - Hanan F Abdul-Rahim
- Department of Public Health, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Gheyath K Nasrallah
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
| | | | - Adeel A Butt
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Hamad Medical Corporation, Doha, Qatar
- Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | | | | | | | | | - Laith J Abu-Raddad
- Infectious Disease Epidemiology Group, Weill Cornell Medicine-Qatar, Cornell University, Qatar Foundation - Education City, Doha, Qatar.
- World Health Organization Collaborating Centre for Disease Epidemiology Analytics on HIV/AIDS, Sexually Transmitted Infections, and Viral Hepatitis, Weill Cornell Medicine-Qatar, Cornell University, Qatar Foundation - Education City, Doha, Qatar.
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Public Health, College of Health Sciences, QU Health, Qatar University, Doha, Qatar.
- College of Health and Life Sciences, Hamad bin Khalifa University, Doha, Qatar.
| |
Collapse
|
44
|
Rubio-Casillas A, Redwan EM, Uversky VN. More antibodies are not always better: Fc effector functions play a critical role in SARS-CoV-2 infection and protection. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:413-447. [PMID: 40246351 DOI: 10.1016/bs.pmbts.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Traditional vaccinology has primarily focused on neutralizing antibody titers as the main correlate of vaccine efficacy, often overlooking the multifaceted roles of antibody Fc effector functions in orchestrating protective immune responses. Fc-mediated immune responses play a pivotal role in immune modulation and pathogen clearance. Emerging evidence from natural infections and vaccine studies highlights the critical contribution of Fc effector functions in determining the quality and durability of immunity. This work explores the limitations of current vaccine evaluation paradigms that prioritize neutralization over Fc effector mechanisms. It also describes findings from a study showing an unexpected role for SARS-CoV-2 anti-spike antibodies: both convalescent plasma and patient-derived monoclonal antibodies (mAbs) lead to maximum phagocytic capacity by monocytes at low concentrations, whereas at higher concentrations the phagocytic capacity was reduced. Given that the severity of COVID-19 disease and antibody titers are strongly positively correlated, this work challenges the paradigm that high antibodies offer better protection against severe disease. It is proposed that humoral and cellular responses elicited by vaccination should never be higher than those produced by natural infection. By integrating antibody Fc effector functions into vaccine development, a paradigm shift is proposed that emphasizes synergic antibody responses. Such an approach could transform vaccine efficacy assessment, enhance protection against dangerous pathogens, and drive innovation in vaccine design.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- Autlan Regional Hospital, Jalisco Health Services, Autlan, Jalisco, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco, Mexico.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg El-Arab, Alexandria, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
45
|
Townsend JP, Hassler HB, Dornburg A. Optimal Annual COVID-19 Vaccine Boosting Dates Following Previous Booster Vaccination or Breakthrough Infection. Clin Infect Dis 2025; 80:316-322. [PMID: 39589144 PMCID: PMC11848277 DOI: 10.1093/cid/ciae559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND COVID-19 booster vaccinations mitigate transmission and reduce the morbidity and mortality associated with infection. However, the optimal date for booster administration remains uncertain. Geographic variation in infection rates throughout the year makes it challenging to intuit the best yearly booster administration date to effectively prevent infection, and also challenging to provide best guidance on how to alter booster administration in response to a breakthrough infection. METHODS We leveraged longitudinal antibody and reinfection probabilities with spatiotemporal projections of COVID-19 incidence to develop a geographically informed approach to optimizing the timing of booster vaccination. We assessed the delay in booster vaccination that is warranted following breakthrough infections whenever they occur during the year, enabling a personalized assessment of optimal timing that acknowledges and respects diversity of COVID-19 immune status, addressing a substantial barrier to uptake. RESULTS Yearly booster vaccination on any date is beneficial to prevention of infection. However, each location exhibits as much as a 3-4-fold range in degree of protection by date of uptake. Optimal COVID-19 booster vaccination dates are location-specific, typically in early autumn in the Northern Hemisphere. Infection late in the interval between boosts substantially alters the optimal boosting date. CONCLUSIONS Considerable benefit accrues from aptly timing COVID-19 booster vaccination campaigns, which can be tailored to specific locations. Individuals can acquire the greatest benefit from booster vaccination by timing it optimally, including delaying in cases of infection late in the interval between boosts. These results provide location-specific guidance for public health policy, healthcare provider recommendations, and individual decision-making.
Collapse
Affiliation(s)
- Jeffrey P Townsend
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, Connecticut, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, USA
- Program in Microbiology, Yale University, New Haven, Connecticut, USA
| | - Hayley B Hassler
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA
- Interdisciplinary Graduate Program in Quantitative Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Alex Dornburg
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| |
Collapse
|
46
|
Effa EE, Ita O, Mwankon J, Siyanbade F, Iwomi F, Ochodo E, Villanueva G, Meremikwu MM. Post-exposure testing at healthcare facilities with SARS-CoV-2 transmission: A rapid review. J Public Health Afr 2025; 16:623. [PMID: 40083354 PMCID: PMC11905177 DOI: 10.4102/jphia.v16i2.623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 11/13/2024] [Indexed: 03/16/2025] Open
Abstract
Background Post-exposure severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) testing following health facility outbreaks may control the spread of infection. Aim This study aimed to assess the impact of testing for SARS-CoV-2 infection on health outcomes during healthcare facility outbreaks. Setting This review included studies conducted at skilled nursing facilities, a cancer centre, and a geriatric psychiatric facility. Methods We followed the methods for conducting rapid systematic reviews, searched databases from December 2019 to August 2022, assessed the risk of bias using the modified Newcastle Ottawa scale, and graded the certainty of evidence using the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) approach. We pooled the prevalence, mortality, and hospitalisation results as appropriate. Results Of the 3055 articles from database search, no study was eligible for inclusion as outlined in the protocol. However, eight non-comparative reports (case series) in skilled nursing facilities were included. The pooled prevalence of SARS-CoV-2 infection among residents of care homes and patients were 38% (95% confidence interval [CI] = 25% - 51%; 5 studies, 2044 participants; I 2 = 94%, very low certainty evidence) and was 12% (95% CI = 6% - 19%; 5 studies, 2312 participants; I 2 = 94%, very low certainty evidence) for exposed healthcare workers. The pooled mortality estimate and hospitalisation rate were 17% and 24%, respectively, (very low certainty evidence). Conclusion There is no identified evidence for or against testing of people in healthcare facilities where there is ongoing transmission of SARS-CoV-2 infection. Contribution The evaluation of the effectiveness of testing strategies during SARS-CoV-2 outbreaks need baseline and follow-up data from well-designed before and after studies appropriate for the setting.
Collapse
Affiliation(s)
- Emmanuel E Effa
- Department of Internal Medicine, Faculty of Clinical Sciences, University of Calabar, Calabar, Nigeria
| | - Okokon Ita
- Department of Medical Microbiology, Faculty of Laboratory Medicine, University of Calabar, Calabar, Nigeria
| | - Joshua Mwankon
- Department of Family Medicine, Faculty of Clinical Sciences, University of Calabar, Calabar, Nigeria
| | - Funmi Siyanbade
- School of Nursing, University of Calabar Teaching Hospital, Calabar, Nigeria
| | - Francis Iwomi
- Cochrane Nigeria, Calabar Institute of Tropical Disease Research and Prevention, University of Calabar Teaching Hospital, Calabar, Nigeria
| | - Eleanor Ochodo
- Centre for Global Health Research, Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | | | - Martin M Meremikwu
- Department of Paediatrics, University of Calabar Teaching Hospital, Calabar, Nigeria
| |
Collapse
|
47
|
Zhao F, Zhang Y, Zhang Z, Chen Z, Wang X, Wang S, Li R, Li Y, Zhang Z, Zheng W, Wang Y, Zhang Z, Wu S, Yang Y, Zhang J, Zai X, Xu J, Chen W. Epitope-focused vaccine immunogens design using tailored horseshoe-shaped scaffold. J Nanobiotechnology 2025; 23:119. [PMID: 39966941 PMCID: PMC11834273 DOI: 10.1186/s12951-025-03200-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
The continuous emergence of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants highlights the need to update coronavirus 2019 disease (COVID-19) vaccine components. Epitope-based vaccine designs targeting conserved and immunorecessive regions of SARS-CoV-2 are critically needed. Here, we report an engineered epitope-focused immunogen design based on a novel horseshoe-shaped natural protein scaffold, named ribonuclease inhibitor 1 (RNH1), that can multiply display of conserved neutralizing epitopes from SARS-CoV-2 S2 stem helix. The designed immunogen RNH1-S1139 demonstrates high binding affinity to S2-specific neutralizing antibodies and elicits robust epitope-targeted antibody responses either through homologous or heterologous vaccination regimens. RNH1-S1139 immune serum has been proven to have similar binding ability against SARS-CoV, SARS-CoV-2 and its variants, providing broad-spectrum protection as a membrane fusion inhibitor. Further studies showed that RNH1 has the potential to serve as a versatile scaffold that displays other helical epitopes from various antigens, including respiratory syncytial virus (RSV) F glycoprotein. Our proposed immunogen engineering strategy via tailored horseshoe-shape nano-scaffold supports the continued development of epitope-focused vaccines as part of a next-generation vaccine design.
Collapse
Affiliation(s)
- Fangxin Zhao
- School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yue Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Zhiling Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Zhengshan Chen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiaolin Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Shaoyan Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Ruihua Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yaohui Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Zhang Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Wanru Zheng
- School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yudong Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Zhe Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Shipo Wu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yilong Yang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Jun Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiaodong Zai
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Junjie Xu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Wei Chen
- School of Medicine, Zhejiang University, Hangzhou, 310058, China.
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China.
- Lead Contact, Beijing, China.
| |
Collapse
|
48
|
Flury P, Krüger N, Sylvester K, Breidenbach J, Al Hamwi G, Qiao J, Chen Y, Rocha C, Serafim MSM, Barbosa da Silva E, Pöhlmann S, Poso A, Kronenberger T, Rox K, O'Donoghue AJ, Yang S, Sträter N, Gütschow M, Laufer SA, Müller CE, Pillaiyar T. Design, Synthesis, and Unprecedented Interactions of Covalent Dipeptide-Based Inhibitors of SARS-CoV-2 Main Protease and Its Variants Displaying Potent Antiviral Activity. J Med Chem 2025; 68:3626-3652. [PMID: 39813204 DOI: 10.1021/acs.jmedchem.4c02254] [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: 01/18/2025]
Abstract
The main protease (Mpro) of SARS-CoV-2 is a key drug target for the development of antiviral therapeutics. Here, we designed and synthesized a series of small-molecule peptidomimetics with various cysteine-reactive electrophiles. Several compounds were identified as potent SARS-CoV-2 Mpro inhibitors, including compounds 8n (IC50 = 0.0752 μM), 8p (IC50 = 0.0887 μM), 8r (IC50 = 0.0199 μM), 10a (IC50 = 0.0376 μM), 10c (IC50 = 0.0177 μM), and 10f (IC50 = 0.0130 μM). Most of them additionally inhibited cathepsin L and were also active against SARS-CoV-1 and MERS-CoV Mpro. In Calu-3 cells, several inhibitors, including 8r, 10a, and 10c, displayed high antiviral activity in the nanomolar range without showing cellular toxicity. The cocrystal structure of SARS-CoV-2 Mpro in complex with 8p revealed covalent binding to the enzyme's catalytic residue Cys145 and showed specific, unprecedented interactions within the substrate binding pocket. Compounds 10c and especially 8n were effective against a panel of naturally occurring nirmatrelvir-resistant mutants, particularly E166V, and showed metabolic stability and additional favorable pharmacokinetic properties, making it a suitable candidate for further preclinical development.
Collapse
Affiliation(s)
- Philipp Flury
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Nadine Krüger
- Platform Infection Models, German Primate Center, Leibniz Institute for Primate Research Göttingen, Kellnerweg 4, 37077 Göttingen, Germany
| | - Katharina Sylvester
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Julian Breidenbach
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Ghazl Al Hamwi
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Jingxin Qiao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Cheila Rocha
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, Göttingen 37077, Germany
| | - Mateus Sá Magalhães Serafim
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0657, United States
| | - Elany Barbosa da Silva
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0657, United States
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, Göttingen 37077, Germany
- Faculty of Biology and Psychology, Georg-August University Göttingen, Göttingen, 37073, Germany
| | - Antti Poso
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio 70211, Finland
| | - Thales Kronenberger
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio 70211, Finland
- German Center for Infection Research (DZIF), Partner Site Tübingen, Elfriede-Aulhorn-Str. 6, Tübingen 72076, Germany
| | - Katharina Rox
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig 38124, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig 38124, Germany
| | - Anthony J O'Donoghue
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0657, United States
| | - Shengyong Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Norbert Sträter
- Center for Biotechnology and Biomedicine, Leipzig University, Deutscher Platz 5, Leipzig 04103, Germany
| | - Michael Gütschow
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Stefan A Laufer
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- Cluster of Excellence "Image Guided and Functionally Instructed Tumor Therapies" (iFIT), Eberhard Karls University of Tuebingen, Tuebingen 72076, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Thanigaimalai Pillaiyar
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| |
Collapse
|
49
|
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
|
50
|
Lin W, Kung KH, Chan CL, Chuang SK, Au KW. Characteristics and risk factors associated with COVID-19 reinfection in Hong Kong: a retrospective cohort study. Epidemiol Infect 2025; 153:e30. [PMID: 39916599 PMCID: PMC11869080 DOI: 10.1017/s0950268825000172] [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: 08/29/2024] [Revised: 12/15/2024] [Accepted: 01/27/2025] [Indexed: 02/28/2025] Open
Abstract
We aimed to identify risk factors related to COVID-19 reinfection in Hong Kong. We performed a population-based retrospective cohort study and reviewed case-based data on COVID-19 infections reported to the Centre for Health Protection from 8 January 2020 to 29 January 2023. We analyzed the epidemiology of COVID-19 infections and performed a Cox regression analysis. In this period, 3.32% (103,065/3,106,579) of COVID-19 infections recorded were classified as reinfection. Compared with primarily infected cases, a higher proportion of re-infected cases had chronic diseases (33.54% vs. 27.27%) and were residents of residential care homes (RCH) (10.99% vs. 1.41%). The time interval between the two episodes ranged from 31 to 1,050 days (median 282 days). Cox regression analysis of Omicron cases with the adjustment of covariates showed that being female (Hazard Ratio [HR] 1.12, 95% CI 1.11-1.13), chronic diseases (HR 1.18, 95% CI 1.16-1.20) and RCH residents (HR 6.78, 95% CI 6.61-6.95) were associated with reinfection, while additional vaccination after primary infection was protective (HR 0.80, 95% CI 0.79-0.81). Further analytical studies on the risk factors and protectors of COVID-19 reinfection are needed to guide targeted interventions.
Collapse
Affiliation(s)
- Wenhua Lin
- Communicable Disease Branch, Centre for Health Protection, Department of Health, Hong Kong
| | - Kin Hang Kung
- Communicable Disease Branch, Centre for Health Protection, Department of Health, Hong Kong
| | - Chung Lam Chan
- Communicable Disease Branch, Centre for Health Protection, Department of Health, Hong Kong
| | - Shuk Kwan Chuang
- Communicable Disease Branch, Centre for Health Protection, Department of Health, Hong Kong
| | - Ka Wing Au
- Communicable Disease Branch, Centre for Health Protection, Department of Health, Hong Kong
| |
Collapse
|