1
|
Zhang L, Yang L, Zhou W, Jian W. The protective effect of emergency fourth-dose vaccination issued to county-level hospital nurses against the Omicron infection peak: evidence from China. BMC Nurs 2025; 24:529. [PMID: 40369491 PMCID: PMC12079848 DOI: 10.1186/s12912-025-03172-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND County-level hospitals are the main providers of health services in rural areas in China. On the eve of the Chinese government's plan to lift the Zero-COVID policy, a number of healthcare workers in county-level hospitals received emergency fourth-dose vaccination against COVID-19. This study aims to evaluate the extent to which these rapid emergency fourth-dose vaccination administered to county-level hospital nurses have provided protection against the Omicron infection wave affecting Mainland China. METHODS A total of 3,302 clinical nurses from 40 county-level hospitals in mainland China participated in this study. The control group was set to comprise nurses who had not received a fourth dose within the past month or indeed any dose of the COVID-19 vaccine within the previous 6 months. The intervention group was set to comprise nurses who received emergency fourth-dose vaccine doses within the month preceding the lifting of the Zero-COVID policy and those who had not received such a fourth-dose within the prior month but who had received a dose of the COVID-19 vaccine within the previous 6 months. Regression methods were used to analyze the factors associated with the probability of symptoms, duration, recovery time and hospitalization rates. RESULTS About 13.1% of the nurses surveyed reported having received the emergency fourth-dose vaccination. It emerged that those nurses had a lower risk of developing clinical symptoms such as fever and diarrhea. Where they did experience symptoms, the duration of these tended to be shorter, with an accompanying and significant reduction in hospitalization rates. It was also found that emergency vaccination was associated with significantly shorter recovery time. CONCLUSIONS The emergency fourth-dose COVID-19 vaccination has had a significant protective effect. At a broader level, reducing hesitancy towards booster shots is an important part of protecting the health of healthcare workers and thereby reducing the impact of the pandemic on the healthcare system and maintaining its resilience.
Collapse
Affiliation(s)
- Lanyue Zhang
- Department of Health Policy and Management, School of Public Health, Peking University, No. 38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Lei Yang
- Henan Honliv Hospital, Changyuan City, Henan Province, 453400, China
| | - Wuping Zhou
- Department of Health Policy and Management, School of Public Health, Peking University, No. 38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Weiyan Jian
- Department of Health Policy and Management, School of Public Health, Peking University, No. 38 Xueyuan Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
2
|
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
|
3
|
Browne DJ, Crooks P, Smith C, Doolan DL. Differential reactivity of SARS-CoV-2 S-protein T-cell epitopes in vaccinated versus naturally infected individuals. Clin Transl Immunology 2025; 14:e70031. [PMID: 40342296 PMCID: PMC12056234 DOI: 10.1002/cti2.70031] [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: 11/18/2024] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 05/11/2025] Open
Abstract
Objectives Vaccine-induced protective immunity against SARS-CoV-2 has proved difficult to sustain. Robust T-cell responses are thought to play an important role, but T-cell responses against the SARS-CoV-2 spike protein (S-protein), the core vaccine antigen, following vaccination or natural infection are incompletely understood. Methods Herein, the reactivity of 170 putative SARS-CoV-2 S-protein CD8+ and CD4+ T-cell peptide epitopes in the same individuals prior to vaccination, after COVID-19 vaccination, and again following subsequent natural infection was assayed using a high-throughput reverse transcription-quantitative PCR (HTS-RT-qPCR) assay. Results The profile of immunoreactive SARS-CoV-2 S-protein epitopes differed between vaccination and natural infection. Vaccine-induced immunoreactive epitopes were localised primarily into two extra-domanial regions. In contrast, epitopes recognised following natural infection were spread across the antigen. Furthermore, T-cell epitopes in naïve individuals were primarily recognised in association with HLA-A, while natural infection shifted epitope associations towards HLA-B, particularly the B7 supertype. Conclusion This study provides insight into T-cell responses against the SARS-CoV-2 S-protein following vaccination and subsequent natural infection.
Collapse
Affiliation(s)
- Daniel J Browne
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsQLDAustralia
| | - Pauline Crooks
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of ImmunologyQIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of ImmunologyQIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
- Faculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
| | - Denise L Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsQLDAustralia
- Institute for Molecular BioscienceThe University of QueenslandSt LuciaQLDAustralia
| |
Collapse
|
4
|
Moore M, Anderson L, Schiffer JT, Matrajt L, Dimitrov D. Durability of COVID-19 vaccine and infection induced immunity: A systematic review and meta-regression analysis. Vaccine 2025; 54:126966. [PMID: 40048931 DOI: 10.1016/j.vaccine.2025.126966] [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: 07/02/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND Despite the success of mRNA vaccines, COVID-19 remains a significant public health threat. Waning of immune memory and the emergence of new variants can degrade population-level protection and contribute to ongoing morbidity. METHODS In this systematic review and meta-regression, we searched for studies in PubMed, medRxiv and bioRxiv published January 1, 2020 - January 1, 2023 measuring vaccine effectiveness as the reduction in infection, symptomatic disease, and severe disease (resulting in hospitalization and/or death) conferred by mRNA-based vaccination and prior SARS-CoV-2 infections relative to naïve individuals. We excluded studies that did not distinguish between mRNA and non-mRNA vaccines or had less than 1000 participants. Using a multi-level model, we quantified the initial effectiveness and change over four to six months following vaccination or infection. Model covariates were COVID variant, number of vaccine doses, and the number and variant of prior infection. Our estimates were adjusted for the age of the study population. FINDINGS Of 828 screened, we included 123 studies in our analysis. Vaccine effectiveness against infection and disease declined both over time and with the emergence of Omicron, regardless of booster doses, though protection against severe outcomes was more durable. Booster doses reduced severe Omicron infections by 90.5 % (95 % confidence interval 87.1-93.8) and 77.6 % (70.5-84.7) at two and 26 weeks post-vaccination, respectively. Protection conferred by hybrid immunity was more durable than that from either vaccination or prior infection alone, but protection against Omicron reinfection was only 50.1 % (32.5-67.8) at 26 weeks following vaccination. Individuals with hybrid immunity had 80.6 % protection (70.0-91.2) following booster doses declining to 36.9 % (19.3-54.6) after 16 weeks. INTERPRETATION Our results suggest that timely deployment of pre-existing boosters can greatly mitigate seasonal COVID outbreaks even in populations with prior infection and vaccination. FUNDING Centers for Disease Control and Prevention (NU38OT000297-03).
Collapse
Affiliation(s)
- Mia Moore
- Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA, USA.
| | - Larissa Anderson
- Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA, USA
| | - Joshua T Schiffer
- Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA, USA
| | - Laura Matrajt
- Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA, USA
| | - Dobromir Dimitrov
- Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA, USA
| |
Collapse
|
5
|
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
|
6
|
Rodriguez-Idiazabal L, Quintana JM, Garcia-Asensio J, Legarreta MJ, Larrea N, Barrio I. Clinically meaningful phenotypes among SARS-CoV-2 reinfections: Informing prevention strategies for future pandemics. Prev Med 2025; 193:108259. [PMID: 40064450 DOI: 10.1016/j.ypmed.2025.108259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
OBJECTIVE Rapidly phenotyping patients can inform public health action plans in new pandemics. This study aimed to derive meaningful SARS-CoV-2 reinfected patients' phenotypes based on easily-available patient data and explore key epidemiological factors of reinfections. METHODS We conducted a retrospective study of a cohort of SARS-CoV-2 reinfected adults from the Basque Country between January 1, 2021 and January 9, 2022. Phenotypes were defined in an unsupervised manner with clustering algorithms, incorporating variables like age, Charlson score, vaccination status and pre-existing treatments and comorbidities. Subsequently, clinical characteristics of phenotypes were compared, and their behavioral differences were evaluated through generalized additive models. Finally, their association with clinical outcomes was assessed. RESULTS Four phenotypes were identified, which subsequently had a direct relationship with the risk levels for severe COVID-19 outcomes. The highest-risk group, phenotype 4, consisted of older adults -76 years, [62-85] (Median, [Interquartile range])- with multiple comorbidities and extensive baseline medication use. Phenotype 3 was slightly younger -64 years, [58-77]- but presented very low Charlson scores and few comorbidities, representing an intermediate-risk group. Phenotypes 1 and 2 were younger and healthier adults with similar clinical profiles. However, phenotype 1 showed a less protective attitude, with a higher rate of unvaccinated patients and shorter time intervals between infections. CONCLUSIONS We were able to classify reinfected patients into four distinct groups based on easily available variables, and these phenotypes had a direct relationship with COVID-19 clinical outcomes. Thus, rapidly phenotyping infected individuals can serve as a preventive public health strategy during new pandemics.
Collapse
Affiliation(s)
- Lander Rodriguez-Idiazabal
- Department of Mathematics, University of the Basque Country UPV/EHU, Leioa, Basque Country, Spain; Applied Statistics Group, Basque Centre for Applied Mathematics (BCAM), Bilbao, Basque Country, Spain; Network for Research on Chronicity, Primary Care, and Health Promotion (RICAPPS), Spain; Biosistemak Institute for Health Systems Research, Barakaldo, Basque Country, Spain.
| | - Jose M Quintana
- Network for Research on Chronicity, Primary Care, and Health Promotion (RICAPPS), Spain; Biosistemak Institute for Health Systems Research, Barakaldo, Basque Country, Spain; Research Unit, Galdakao-Usansolo University Hospital, Osakidetza Basque Health Service, Galdakao, Basque Country, Spain; Health Service Research Network on Chronic Diseases (REDISSEC), Bilbao, Basque Country, Spain.
| | - Julia Garcia-Asensio
- Office of Healthcare Planning, Organization and Evaluation, Basque Government Department of Health, Basque Country, Spain.
| | - Maria Jose Legarreta
- Network for Research on Chronicity, Primary Care, and Health Promotion (RICAPPS), Spain; Biosistemak Institute for Health Systems Research, Barakaldo, Basque Country, Spain; Research Unit, Galdakao-Usansolo University Hospital, Osakidetza Basque Health Service, Galdakao, Basque Country, Spain; Health Service Research Network on Chronic Diseases (REDISSEC), Bilbao, Basque Country, Spain.
| | - Nere Larrea
- Network for Research on Chronicity, Primary Care, and Health Promotion (RICAPPS), Spain; Biosistemak Institute for Health Systems Research, Barakaldo, Basque Country, Spain; Research Unit, Galdakao-Usansolo University Hospital, Osakidetza Basque Health Service, Galdakao, Basque Country, Spain; Health Service Research Network on Chronic Diseases (REDISSEC), Bilbao, Basque Country, Spain.
| | - Irantzu Barrio
- Department of Mathematics, University of the Basque Country UPV/EHU, Leioa, Basque Country, Spain; Applied Statistics Group, Basque Centre for Applied Mathematics (BCAM), Bilbao, Basque Country, Spain.
| |
Collapse
|
7
|
Nouanesengsy A, Semesi A, Quach K, Ivanochko D, Byrne W, Hwang M, La Neve MR, Leon-Ponte M, Litosh A, Wisener N, Adeli K, Campigotto A, Grunebaum E, McGeer A, Moraes TJ, Sepiashvili L, Upton J, Julien JP, Allen U. Persistence and decay of neutralizing antibody responses elicited by SARS-CoV-2 infection and hybrid immunity in a Canadian cohort. Microbiol Spectr 2025; 13:e0133324. [PMID: 39969224 PMCID: PMC11960127 DOI: 10.1128/spectrum.01333-24] [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: 06/19/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
A major challenge with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has been assessing the intensity, dynamics, and determinants of the antibody responses after infection and/or vaccination. Therefore, we aimed to characterize the longitudinal dynamics of the antibody responses among naturally infected individuals and individuals who achieved hybrid immunity in a large Canadian cohort. We demonstrate that anti-Spike IgGs and neutralizing antibody dynamics vary greatly among individuals with COVID-19, in peak antibody levels, rate of waning, and longevity of the antibody response. Additionally, we found an association between robust antibody responses and individuals with severe COVID-19 clinical symptoms during the first-month post-symptom onset. For individuals who achieved hybrid immunity, a robust increase in anti-S1 IgGs and neutralizing antibodies followed the first vaccination dose; however, there was a minimal increase in the anti-S1 IgGs and neutralizing antibody titers after administration of the second dose of the vaccine. Furthermore, neutralizing antibodies elicited by the wild-type virus alone were largely ineffective against emerging variants of concern in our natural infection-only cohort, in contrast to a much broader and more robust neutralization profile observed in individuals who achieved hybrid immunity. Our findings emphasize the need for global SARS-CoV-2 vaccination efforts to further sustain protective immune responses required to minimize viral spread and disease severity in the population. As SARS-CoV-2 variants continue to emerge, understanding the interplay between previous infections, vaccine durability, and virus evolution will be critical for guiding ongoing vaccination strategies. IMPORTANCE A major challenge with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has been assessing the intensity, dynamics, and determinants of the antibody response after infection and/or vaccination. Our paper addresses this in a large Canadian cohort with antibody responses that were generated by natural infection as well as vaccine in some persons studied.
Collapse
Affiliation(s)
- Amy Nouanesengsy
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Anthony Semesi
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
| | - Kim Quach
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Danton Ivanochko
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
| | - Walter Byrne
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Matthew Hwang
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maria-Rosa La Neve
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Matilde Leon-Ponte
- Division of Allergy and Immunology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alice Litosh
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nicole Wisener
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Khosrow Adeli
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Aaron Campigotto
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Eyal Grunebaum
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
| | - Allison McGeer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tunenbaum Research Institute at Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Theo J. Moraes
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lusia Sepiashvili
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Julia Upton
- Division of Allergy and Immunology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Upton Allen
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
- Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Fant P, Laurent S, Desert P, Combadière B, Palazzi X, Choudhary S, Gervais F, Broudic K, Rossi R, Gauthier BE. Proceedings of the 2023 Annual Scientific Meeting of the French Society of Toxicologic Pathology (SFPT) on Preclinical Development and Therapeutic Applications of mRNA-Based Technologies. Toxicol Pathol 2025:1926233251326089. [PMID: 40110665 DOI: 10.1177/01926233251326089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
The 2023 annual scientific meeting of the French Society of Toxicologic Pathology (Société Française de Pathologie Toxicologique, SFPT), entitled "mRNA-based technologies: preclinical development and therapeutic applications," was held in Lyon (France) on May 25 to 26, 2023. The aim of the meeting was to discuss the biology, immunology, and preclinical development of messenger RNA (mRNA)-based vaccines and therapeutics, including immuno-oncology and rare diseases, as well as the regulatory aspect of the COVID-19 vaccines and an overview of the principles and applications of in situ hybridization techniques. This article presents the summary of five lectures along with selected figures, tables, and key literature references on this topic.
Collapse
Affiliation(s)
- Pierluigi Fant
- Charles River Laboratories Safety Assessment, Saint Germain-Nuelles, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Sikuvi K, Nghitukwa N, Kakehongo N, Katjitae I, Matos C, Oedi P, Netha SM, Nepolo E, Winter C. Effectiveness of COVID-19 vaccines against laboratory-confirmed SARS-CoV-2 infection amongst health workers, Windhoek, Namibia. Vaccine 2025:126977. [PMID: 40074602 DOI: 10.1016/j.vaccine.2025.126977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025]
Abstract
INTRODUCTION As of 24 October 2021, 128,868 laboratory-confirmed COVID-19 cases and 3550 deaths were reported from Namibia. The national COVID-19 vaccination campaign that started in March 2021 included health workers (HWs) as a priority group. The vaccines most administered were Sinopharm, AstraZeneca, Pfizer-BioNtech, and Janssen. We aimed to measure the effectiveness of COVID-19 vaccines (VE) amongst HWs against laboratory-confirmed SARS-CoV-2 infection in Namibia. METHODS We conducted a test negative design (TND) amongst HWs from the two main hospitals treating COVID-19 patients. HWs were defined as all hospital staff over 18 years in direct or indirect contact with patients, eligible for COVID-19 vaccination. We interviewed actively recruited HWs with standardized questionnaires in-person from 25/10/2021 to 25/4/2022. The participants had to state their vaccination status, which was verified through vaccination card, vaccine registry and/or District Health Information System 2. RT-PCR testing of respiratory specimens and serological testing (Wantai and Platelia-ELISA) were conducted. We measured VE by comparing the vaccination status between RT-PCR positive and negative HWs using a multivariable logistic regression model, which was adjusted for confounders. We calculated VE = (1-odds ratio of vaccination)*100 %. RESULTS We included 1201 HWs of which 322 (26.8 %) participants were fully vaccinated with a primary series against COVID-19, 62 (5.2 %) were partially vaccinated and 735 (61.2 %) were not vaccinated. In total, 1119 (93 %) participants had antibodies against SARS-CoV-2 including 637 (90 %) of the unvaccinated participants. Fifty-eight (4.8 %) participants tested RT-PCR positive for SARS-CoV-2. The Omicron variant was detected in all 13 sequenced genomes (11 BA.1.18, 2 BA.1). The estimated overall VE for full vaccination was 61.8 % (95 %-confidence interval, 9.3-83.9 %). CONCLUSIONS The VE results suggest that COVID-19 vaccines used in Namibia provided good protection from infections with the Omicron variant even if many participants had a SARS-CoV-2 infection before the study. Therefore, COVID-19 vaccines should be administered to risk groups such as HWs independent from previous infections.
Collapse
Affiliation(s)
- Kaveto Sikuvi
- Ministry of Health and Social Services Namibia, Ministerial Building, Harvey Street, Windhoek, Namibia
| | - Natasha Nghitukwa
- Ministry of Health and Social Services Namibia, Ministerial Building, Harvey Street, Windhoek, Namibia
| | - Ndiitodino Kakehongo
- Department of Medical Sciences, School of Medicine, University of Namibia, Windhoek, Namibia
| | - Ismael Katjitae
- Ministry of Health and Social Services Namibia, Ministerial Building, Harvey Street, Windhoek, Namibia
| | - Carolina Matos
- Robert Koch Institute, Am Nordufer 20, 13353 Berlin, Germany
| | - Philip Oedi
- Robert Koch Institute, Am Nordufer 20, 13353 Berlin, Germany
| | - Sibongile Manga Netha
- Department of Human, Biological and Translational Medical Science, School of Medicine, University of Namibia, Windhoek, Namibia
| | - Emmanuel Nepolo
- Department of Human, Biological and Translational Medical Science, School of Medicine, University of Namibia, Windhoek, Namibia
| | | |
Collapse
|
10
|
Dodge MC, Ellervik C, Kataria Y. A Meta-analysis of Severe Acute Respiratory Syndrome Coronavirus 2 Anti-spike Immunoglobulin G Antibody Durability up to 9 Months After Full Vaccination in Adults. Clin Lab Med 2025; 45:111-136. [PMID: 39892931 DOI: 10.1016/j.cll.2024.10.007] [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: 02/04/2025]
Abstract
This meta-analysis aims to estimate temporal decline in vaccine-induced antibodies against severe acute respiratory syndrome coronavirus 2 up to 9 months after full vaccination contributing to overall understanding of coronavirus disease 2019 infection protection. We identified 15 eligible studies and calculated standardized mean differences (SMD) between antibody concentrations at 1, 3, 6, and 9 months after full vaccination. Overall SMD between 1 month after vaccination and 3 months was -1.14 (95% CI -1.52, -0.76), at 6 months was -1.06 (95% CI -1.30, -0.81), and at 9 months, it was -0.77 (95% CI -0.94, -0.60) suggesting a moderate decline over time.
Collapse
Affiliation(s)
- Maura C Dodge
- Department of Pathology and Laboratory Medicine, Boston Medical Center, 670 Albany Street, 6th Floor, Boston, MA 02118, USA
| | - Christina Ellervik
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; Department of Laboratory Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Pathology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Yachana Kataria
- Department of Pathology and Laboratory Medicine, Boston Medical Center, 670 Albany Street, 6th Floor, Boston, MA 02118, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, 670 Albany Street, 4th Floor, Boston, MA 02118, USA; Clinical Chemistry, Department of Pathology and Laboratory Medicine, Boston Medical Center, 670 Albany Street, Boston, MA 02118, USA.
| |
Collapse
|
11
|
Wang Y, So HC, Tsang NNY, Kwok SK, Cowling BJ, Leung GM, Ip DKM. Clinical profile analysis of SARS-CoV-2 community infections during periods with omicron BA.2, BA.4/5, and XBB dominance in Hong Kong: a prospective cohort study. THE LANCET. INFECTIOUS DISEASES 2025; 25:276-289. [PMID: 39419049 DOI: 10.1016/s1473-3099(24)00574-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Existing studies on SARS-CoV-2 infection have mainly focused on severe clinical outcomes; understanding of the clinical severity profile of general community infections is poor. We aimed to assess and compare the clinical profiles of infections with SARS-CoV-2 omicron (B.1.1.529) subvariants in a representative community cohort in Hong Kong during periods of BA.2, BA.4/5, and XBB dominance. METHODS In this prospective cohort study in Hong Kong, a representative community cohort of individuals aged at least 5 years were recruited by random-digit dialling and underwent weekly rapid antigen testing for SARS-CoV-2, irrespective of symptoms, during three periods from March 1, 2022, to Oct 31, 2023, in which the BA.2, BA.4/5, or XBB subvariants were dominant. We analysed the likelihood of symptoms, as well as the patterns, severity, and duration of symptoms and their associations with participant demographics and vaccination and infection histories. FINDINGS 1126 (11·0%) of 10 279 participants in the BA.2 period, 830 (6·6%) of 12 588 in the BA.4/5 period, and 633 (11·1%) of 5690 during the XBB period tested positive for SARS-CoV-2 infection on rapid antigen tests. Community infections were generally mild, with asymptomatic infections comprising 22·0-25·0% of infections. No hospitalisations or deaths occurred as a direct result of SARS-CoV-2 infection during the study period. Compared with children aged 5-17 years, a higher likelihood of being symptomatic on infection was found for adults aged 18-59 years during the period of BA.2 dominance and adults aged 60 years or older during XBB dominance. Most (>90·0%) participants with symptomatic infections reported respiratory and systemic symptoms. Up-to-date vaccination with a regimen containing the BNT162b2 vaccine, compared with those without an up-to-date vaccine, was associated with a reduced likelihood of symptoms on infection during the period of BA.2 dominance and of severe symptoms causing substantial disturbance to daily life (grade 3 symptoms) during periods of BA.2 and BA.4/5 dominance, whereas no association was observed during the period of XBB dominance. Previous SARS-CoV-2 infection was associated with a reduced likelihood of symptoms on infection during BA.4/5 and XBB dominance and of severe symptoms during XBB dominance. Reports of severe symptoms increased over the three periods, from 236 (27·7%) of 852 symptomatic participants during BA.2 dominance to 176 (37·1%) of 475 during XBB dominance. The duration of symptoms was longest in the BA.2 period (median 10·0 days [95% CI 9·0-10·0]) and similar in the other two periods (8·0 [8·0-9·0] during BA.4/5 dominance and 8·0 [8·0-9·0] during XBB dominance). A symptom duration of 60 days or longer was reported only during the period of BA.2 dominance, in six (0·7%) of 824 infections. INTERPRETATION SARS-CoV-2 infections were generally mild, but not increasingly so, along the evolution of omicron subvariants in this highly vaccinated population. About a third of participants with symptomatic infections reported that the symptoms severely affected daily life even if they were not admitted to hospital, resulting in morbidity, absence from work or school due to illness, productivity loss, and increased medicoeconomic burden. A gradual reduction in the association of vaccines and increase in the association of previous infection with the symptom profile, possibly reflecting the effects of immune escape and waning, were observed over the study period. FUNDING Henry Fok Foundation and Hong Kong Health Bureau.
Collapse
Affiliation(s)
- Yawei Wang
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Hau Chi So
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Nicole Ngai Yung Tsang
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Siu Kan Kwok
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Benjamin J Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China; Laboratory of Data Discovery for Health, Hong Kong Special Administrative Region, China
| | - Gabriel M Leung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China; Laboratory of Data Discovery for Health, Hong Kong Special Administrative Region, China
| | - Dennis Kai Ming Ip
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
12
|
Mao Y, Guo A, Zhang Y, Lai J, Yuan D, Zhang H, Diao W, Chen W, Yan F. Baricitinib treatment for hospitalized patients with severe COVID-19 on invasive mechanical ventilation: a propensity score-matched and retrospective analysis. Front Med (Lausanne) 2025; 12:1445809. [PMID: 39911872 PMCID: PMC11794113 DOI: 10.3389/fmed.2025.1445809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
Introduction Baricitinib is a selective inhibitor of Janus kinase (JAK)1 and JAK2, which is associated with clinical improvement in non-severe COVID-19 patients. But in severe COVID-19 patients, the effectiveness of baricitinib is still controversial. Methods A propensity score-matched and retrospective study was conducted to evaluate the effectiveness of baricitinib in severe COVID-19 patients requiring invasive mechanical ventilation (IMV). Results A total number of 48 patients treated with baricitinib were included, and 48 patients were assigned to control group by propensity score matching. The mean ages were high in both group (baricitinib group vs. control group: 78.80 ± 9.04 vs. 82.57 ± 9.27), and most were unvaccinated (62.5% vs. 66.7%. Baricitinib group had a higher proportion of patients with hypertension (73.9% vs. 45.5%, p = 0.006). Control group had higher level of creatine kinase-myocardial band (247.50 vs. 104.50, p = 0.021). Patients in the baricitinib group were more likely to receive nirmatrelvir/ritonavir (39.6% vs. 16.7%, p = 0.017) and intravenous immunoglobin (14.6% vs. 0, p = 0.007). Baricitinib group had significantly lower all-cause 28-days mortality than control group (72.9% vs. 89.6%, p = 0.004). Conclusion The present study revealed baricitinib reduced 28-days mortality in severe COVID-19 patients on IMV. The effectiveness of baricitinib in treating patients with severe COVID-19 on IMV needs to be further investigated through future studies.
Collapse
Affiliation(s)
- Yanxiong Mao
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Anyi Guo
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying Zhang
- Department of Scientific Research, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jianxing Lai
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dian Yuan
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hao Zhang
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenqi Diao
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Weisong Chen
- Department of Respiratory, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, China
| | - Fugui Yan
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
13
|
Rankin N, Saiyed S, Du H, Gardner LM. A multi-city COVID-19 forecasting model utilizing wastewater-based epidemiology. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 960:178172. [PMID: 39765166 DOI: 10.1016/j.scitotenv.2024.178172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/23/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
The COVID-19 pandemic highlighted shortcomings in forecasting models, such as unreliable inputs/outputs and poor performance at critical points. As COVID-19 remains a threat, it is imperative to improve current forecasting approaches by incorporating reliable data and alternative forecasting targets to better inform decision-makers. Wastewater-based epidemiology (WBE) has emerged as a viable method to track COVID-19 transmission, offering a more reliable metric than reported cases for forecasting critical outcomes like hospitalizations. Recognizing the natural alignment of wastewater systems with city structures, ideal for leveraging WBE data, this study introduces a multi-city, wastewater-based forecasting model to categorically predict COVID-19 hospitalizations. Using hospitalization and COVID-19 wastewater data for six US cities, accompanied by other epidemiological variables, we develop a Generalized Additive Model (GAM) to generate two categorization types. The Hospitaization Capacity Risk Categorization (HCR) predicts the burden on the healthcare system based on the number of available hospital beds in a city. The Hospitalization Rate Trend (HRT) Categorization predicts the trajectory of this burden based on the growth rate of COVID-19 hospitalizations. Using these categorical thresholds, we create probabilistic forecasts to retrospectively predict the risk and trend category of six cities over a 20-month period for 1, 2, and 3 week forecasting windows. We also propose a new methodology to measure forecasting model performance at change points, or time periods where sudden changes in outbreak dynamics occurred. We also explore the influence of wastewater as a predictor for hospitalizations, showing its inclusion positively impacts the model's performance. With this categorical forecasting study, we are able to predict hospital capacity risk and disease trends in a novel and useful way, giving city decision-makers a new tool to predict COVID-19 hospitalizations.
Collapse
Affiliation(s)
- Naomi Rankin
- Department of Civil and Systems Engineering, Johns Hopkins University, Baltimore, MD, USA.
| | - Samee Saiyed
- Department of Civil and Systems Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Hongru Du
- Department of Civil and Systems Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Lauren M Gardner
- Department of Civil and Systems Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
14
|
Sanin KI, Khanam M, Sharaque AR, Elahi M, Roy BR, Hasan MK, Dutta GK, Dutta A, Islam MN, Islam MS, Khan MNA, Mahmud M, Nadia N, Noushin F, Roy AK, Sarker P, Tofail F. Comparing Antibody Responses to Homologous vs. Heterologous COVID-19 Vaccination: A Cross-Sectional Analysis in an Urban Bangladeshi Population. Vaccines (Basel) 2025; 13:67. [PMID: 39852846 PMCID: PMC11768507 DOI: 10.3390/vaccines13010067] [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: 11/03/2024] [Revised: 12/21/2024] [Accepted: 12/25/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Vaccination has played a crucial role in mitigating the spread of COVID-19 and reducing its severe outcomes. While over 90% of Bangladesh's population has received at least one COVID-19 vaccine dose, the comparative effectiveness of homologous versus heterologous booster strategies, along with the complex interplay of factors within the population, remains understudied. This study aimed to compare antibody responses between these booster approaches. METHODS This cross-sectional study enrolled 723 adults in urban Dhaka who had received COVID-19 booster doses within the last six months. Participants were grouped based on homologous or heterologous booster vaccination. Data were collected through structured household surveys, and 2 mL blood samples were collected for measuring antibody titers. RESULTS Heterologous booster recipients showed higher median antibody titers (8597.0 U/mL, IQR 5053.0-15,482.3) compared to homologous recipients (6958.0 U/mL, IQR 3974.0-12,728.5). In the adjusted analysis, the type of booster dose had no significant impact on antibody levels. However, the duration since the last booster dose was significantly associated with antibody levels, where each additional month since receiving the booster corresponded to approximately a 15-16% reduction in antibody levels (Adj. coeff: 0.85, 95% CI: 0.81, 0.88; p < 0.001). Participants over 40 years demonstrated higher antibody levels than younger individuals (Adj. coeff: 1.23, 95% CI: 1.07, 1.43; p = 0.005). Sex, BMI, and prior COVID-19 infection showed no significant associations with antibody levels after adjustment. CONCLUSION The results underscore the complexity of immune responses across different demographic groups and suggest potential benefits of ongoing heterologous booster strategies in sustaining immunity.
Collapse
Affiliation(s)
- Kazi Istiaque Sanin
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68, Shaheed Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh; (M.K.); (A.R.S.); (M.E.); (B.R.R.); (M.K.H.); (G.K.D.); (A.D.); (A.K.R.); (P.S.); (F.T.)
| | - Mansura Khanam
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68, Shaheed Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh; (M.K.); (A.R.S.); (M.E.); (B.R.R.); (M.K.H.); (G.K.D.); (A.D.); (A.K.R.); (P.S.); (F.T.)
| | - Azizur Rahman Sharaque
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68, Shaheed Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh; (M.K.); (A.R.S.); (M.E.); (B.R.R.); (M.K.H.); (G.K.D.); (A.D.); (A.K.R.); (P.S.); (F.T.)
| | - Mahbub Elahi
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68, Shaheed Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh; (M.K.); (A.R.S.); (M.E.); (B.R.R.); (M.K.H.); (G.K.D.); (A.D.); (A.K.R.); (P.S.); (F.T.)
| | - Bharati Rani Roy
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68, Shaheed Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh; (M.K.); (A.R.S.); (M.E.); (B.R.R.); (M.K.H.); (G.K.D.); (A.D.); (A.K.R.); (P.S.); (F.T.)
| | - Md. Khaledul Hasan
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68, Shaheed Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh; (M.K.); (A.R.S.); (M.E.); (B.R.R.); (M.K.H.); (G.K.D.); (A.D.); (A.K.R.); (P.S.); (F.T.)
| | - Goutam Kumar Dutta
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68, Shaheed Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh; (M.K.); (A.R.S.); (M.E.); (B.R.R.); (M.K.H.); (G.K.D.); (A.D.); (A.K.R.); (P.S.); (F.T.)
| | - Abir Dutta
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68, Shaheed Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh; (M.K.); (A.R.S.); (M.E.); (B.R.R.); (M.K.H.); (G.K.D.); (A.D.); (A.K.R.); (P.S.); (F.T.)
| | - Md. Nazmul Islam
- Communicable Disease Control (CDC), Directorate General of Health Services, Ministry of Health and Family Welfare, Dhaka 1212, Bangladesh; (M.N.I.); (M.S.I.); (M.N.A.K.); (M.M.); (N.N.); (F.N.)
| | - Md. Safiqul Islam
- Communicable Disease Control (CDC), Directorate General of Health Services, Ministry of Health and Family Welfare, Dhaka 1212, Bangladesh; (M.N.I.); (M.S.I.); (M.N.A.K.); (M.M.); (N.N.); (F.N.)
| | - Md. Nasir Ahmed Khan
- Communicable Disease Control (CDC), Directorate General of Health Services, Ministry of Health and Family Welfare, Dhaka 1212, Bangladesh; (M.N.I.); (M.S.I.); (M.N.A.K.); (M.M.); (N.N.); (F.N.)
| | - Mustufa Mahmud
- Communicable Disease Control (CDC), Directorate General of Health Services, Ministry of Health and Family Welfare, Dhaka 1212, Bangladesh; (M.N.I.); (M.S.I.); (M.N.A.K.); (M.M.); (N.N.); (F.N.)
| | - Nuzhat Nadia
- Communicable Disease Control (CDC), Directorate General of Health Services, Ministry of Health and Family Welfare, Dhaka 1212, Bangladesh; (M.N.I.); (M.S.I.); (M.N.A.K.); (M.M.); (N.N.); (F.N.)
| | - Fablina Noushin
- Communicable Disease Control (CDC), Directorate General of Health Services, Ministry of Health and Family Welfare, Dhaka 1212, Bangladesh; (M.N.I.); (M.S.I.); (M.N.A.K.); (M.M.); (N.N.); (F.N.)
| | - Anjan Kumar Roy
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68, Shaheed Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh; (M.K.); (A.R.S.); (M.E.); (B.R.R.); (M.K.H.); (G.K.D.); (A.D.); (A.K.R.); (P.S.); (F.T.)
| | - Protim Sarker
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68, Shaheed Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh; (M.K.); (A.R.S.); (M.E.); (B.R.R.); (M.K.H.); (G.K.D.); (A.D.); (A.K.R.); (P.S.); (F.T.)
| | - Fahmida Tofail
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68, Shaheed Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh; (M.K.); (A.R.S.); (M.E.); (B.R.R.); (M.K.H.); (G.K.D.); (A.D.); (A.K.R.); (P.S.); (F.T.)
| |
Collapse
|
15
|
Edwards CT, Karunakaran KA, Garcia E, Beutler N, Gagne M, Golden N, Aoued H, Pellegrini KL, Burnett MR, Honeycutt CC, Lapp SA, Ton T, Lin MC, Metz A, Bombin A, Goff K, Scheuermann SE, Wilkes A, Wood JS, Ehnert S, Weissman S, Curran EH, Roy M, Dessasau E, Paiardini M, Upadhyay AA, Moore IN, Maness NJ, Douek DC, Piantadosi A, Andrabi R, Rogers TR, Burton DR, Bosinger SE. Passive infusion of an S2-Stem broadly neutralizing antibody protects against SARS-CoV-2 infection and lower airway inflammation in rhesus macaques. PLoS Pathog 2025; 21:e1012456. [PMID: 39847599 PMCID: PMC11793774 DOI: 10.1371/journal.ppat.1012456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 02/04/2025] [Accepted: 12/27/2024] [Indexed: 01/25/2025] Open
Abstract
The continued evolution of SARS-CoV-2 variants capable of subverting vaccine and infection-induced immunity suggests the advantage of a broadly protective vaccine against betacoronaviruses (β-CoVs). Recent studies have isolated monoclonal antibodies (mAbs) from SARS-CoV-2 recovered-vaccinated donors capable of neutralizing many variants of SARS-CoV-2 and other β-CoVs. Many of these mAbs target the conserved S2 stem region of the SARS-CoV-2 spike protein, rather than the receptor binding domain contained within S1 primarily targeted by current SARS-CoV-2 vaccines. One of these S2-directed mAbs, CC40.8, has demonstrated protective efficacy in small animal models against SARS-CoV-2 challenge. As the next step in the pre-clinical testing of S2-directed antibodies as a strategy to protect from SARS-CoV-2 infection, we evaluated the in vivo efficacy of CC40.8 in a clinically relevant non-human primate model by conducting passive antibody transfer to rhesus macaques (RM) followed by SARS-CoV-2 challenge. CC40.8 mAb was intravenously infused at 10mg/kg, 1mg/kg, or 0.1 mg/kg into groups (n = 6) of RM, alongside one group that received a control antibody (PGT121). Viral loads in the lower airway were significantly reduced in animals receiving higher doses of CC40.8. We observed a significant reduction in inflammatory cytokines and macrophages within the lower airway of animals infused with 10mg/kg and 1mg/kg doses of CC40.8. Viral genome sequencing demonstrated a lack of escape mutations in the CC40.8 epitope. Collectively, these data demonstrate the protective efficiency of broadly neutralizing S2-targeting antibodies against SARS-CoV-2 infection within the lower airway while providing critical preclinical work necessary for the development of pan-β-CoV vaccines.
Collapse
Affiliation(s)
- Christopher T. Edwards
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Kirti A. Karunakaran
- Department of Pathology, Microbiology & Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States of America
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Matthew Gagne
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nadia Golden
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Hadj Aoued
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Kathryn L. Pellegrini
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Matthew R. Burnett
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Christopher Cole Honeycutt
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Stacey A. Lapp
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Thang Ton
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Mark C. Lin
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Amanda Metz
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Andrei Bombin
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kelly Goff
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Sarah E. Scheuermann
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Amelia Wilkes
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Jennifer S. Wood
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Stephanie Ehnert
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Stacey Weissman
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Elizabeth H. Curran
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Melissa Roy
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Evan Dessasau
- Division of Histology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Amit A. Upadhyay
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Ian N. Moore
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Nicholas J. Maness
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Daniel C. Douek
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anne Piantadosi
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, California, United States of America
| | - Thomas R. Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, California, United States of America
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, United States of America
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
16
|
Arévalo‐Herrera M, Rincón‐Orozco B, González‐Escobar JM, Herrera‐Arévalo SM, Carrasquilla‐Agudelo E, Serna‐Ortega PA, Quiceno‐García S, Palacio‐Muñoz N, Rosero‐López B, Mondol‐Miranda E, Freyle‐Roman I, Mendoza‐Landinez B, Mora‐Guevara E, Santos‐Barbosa JC, Bohórquez‐Martínez F, Bolaños‐Cristancho N, Jiménez‐Serna M, Nieto‐Rojas MA, Suarez‐Zamora D, Quintero‐Espinosa J, Londoño‐Trujillo D, Herrera‐ Valencia S. Longitudinal Follow-Up of the Specific Antibody Response to SARS-CoV-2 Vaccination in Colombia. J Med Virol 2025; 97:e70133. [PMID: 39817585 PMCID: PMC11737005 DOI: 10.1002/jmv.70133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 11/04/2024] [Accepted: 12/06/2024] [Indexed: 01/18/2025]
Abstract
A total of 5011 adult volunteers attending vaccination centers in different regions of Colombia were enrolled in a 1-year prospective observational cohort study to evaluate the immunogenicity and effectiveness of SARS-CoV-2-based vaccines as part of a National Vaccine Program established to contain the COVID-19 pandemic. Following informed consent, 5,011 participants underwent a sociodemographic survey and PCR testing to assess SARS-CoV-2 infection. Blood samples were collected, and serum fractions were obtained from a participant subsample (n = 3441) at six-time points to assess virus-specific IgG responses to the Spike protein, its Receptor Binding Domain, and the Nucleoprotein by ELISA. Additionally, antibody-neutralizing activity was evaluated using a cPass SARS-CoV-2 neutralization kit. Most participants (95.8%; n = 4802) received between one Ad26. COV2.S (Janssen vaccine) and four vaccine doses of BNT162b2 (Pfizer/BioNTech), AZD1222 (AstraZeneca), mRNA-1273 (Moderna), CoronaVac (Sinovac), with some receiving vaccine combinations; a small group, 4.2% (n = 209), remained unvaccinated. Throughout the study, only 8.76% (n = 439) of the participants tested positive for SARS-CoV-2 by PCR. Notably, all participants seroconverted for IgG antibodies, with high seropositivity rates for S (99.8%; n = 4795), RBD (99.7%; n = 1691), and N (92.7%; n = 3072) proteins. Moreover, significant (92%-97%) neutralizing activity was observed for all four SARS-CoV-2 circulating variants. This study highlights the importance of assessing the duration of the IgG response to SARS-CoV-2 elicited by vaccination and infection, and the antibody neutralizing activity as a potential surrogate marker of protection. These findings provide important insight for further strengthening the vaccination strategies to control COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Juliana Quintero‐Espinosa
- Fundación Santa Fe de BogotáSalud PoblacionalColombia
- Departamento de Medicina Interna, Sección de Infectología, Fundación Santa Fe de Bogotá
| | | | | |
Collapse
|
17
|
Kolomba BM, Kalenga Luhembwe F, Ndala DBB, Kanku Wa Ilunga P, Ciamala Mukendi P, Ngongo Kitenge A, Ngoy Lumbule J, Kilolo Ngoy E, Umba Ilunga A, Mbidi Miema J, Mwavita CK, Mwamba GN, Wa Bene AC, Wakamba AM, Ngongo AN, Kabamba Nzaji M. Healthcare workers' willingness to receive COVID-19 booster dose and associated factors in the Democratic Republic of the Congo. Hum Vaccin Immunother 2024; 20:2357214. [PMID: 38783665 PMCID: PMC11135840 DOI: 10.1080/21645515.2024.2357214] [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: 11/10/2023] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
The COVID-19 booster dose is considered an important adjunct for the control of the COVID-19 pandemic due to reports of reduced immunity in fully vaccinated individuals. The aims of this study were to assess healthcare workers' intention to receive the booster dose of COVID-19 vaccine and to identify predictive factors among healthcare workers. A cross-sectional study was conducted among healthcare workers selected in two provinces, Kasai Oriental, and Haut-Lomami. Data were collected using a questionnaire administered through structured face-to-face interviews, with respondents using a pre-tested questionnaire set up on the Open Data Kit (ODK Collect). All data were analyzed using SPSS v26.0 (IBM Corporation, Armonk, NY, USA). Vaccination coverage for COVID-19, considering declarations by health workers, is around 85.9% for the province of Kasai Oriental and 85.8% for Haut-Lomami. A total of 975 responses were collected, 71.4% of health workers at Kasai Oriental and 66.4% from Haut-Lomami declared a definite willingness to receive a COVID-19 vaccine booster. The duration of protection was the main reason for accepting a booster COVID-19 dose for 64.6% of the respondents. Logistic regression analysis showed that having chronic diseases (aOR = 2.95 [1.65-5.28]), having already received one of the COVID-19 vaccines (aOR = 2.72 [1.43-5. 19]); the belief that only high-risk individuals, such as healthcare professionals and elderly people suffering from other illnesses, needed a booster dose (aOR = 1.75 [1.10-2.81]). Considering the burden of COVID-19, a high acceptance rate for booster doses could be essential to control the pandemic. Our results are novel and could help policymakers design and implement specific COVID-19 vaccination programs to reduce reluctance to seek booster vaccination.
Collapse
Affiliation(s)
- Bertin Mindje Kolomba
- Pediatric Department, Faculty of Medicine, University of Kamina, Kamina, Democratic Republic of the Congo
| | | | - Deca Blood Banza Ndala
- Department of Epidemiology and Public Health, Nursing Care Section, Higher Institute of Medical Techniques of Lubumbashi, Lubumbashi, Democratic Republic of the Congo
| | | | - Paul Ciamala Mukendi
- Department of Teaching and Administration in Nursing, Nursing Section, Higher Institute of Medical Techniques of Mbuji-Mayi, Mbuji-Mayi, Democratic Republic of the Congo
| | - Amide Ngongo Kitenge
- School of Public Health, University of Kamina, Kamina, Democratic Republic of the Congo
| | - John Ngoy Lumbule
- School of Public Health, University of Kamina, Kamina, Democratic Republic of the Congo
| | - Elie Kilolo Ngoy
- Department of Public Health, Faculty of Medicine, University of Kamina, Kamina, Democratic Republic of the Congo
| | - Antoine Umba Ilunga
- Department of Internal Medicine, Faculty of Medicine, University of Kamina, Kamina, Democratic Republic of the Congo
| | - Judith Mbidi Miema
- Pediatric Department, Faculty of Medicine, University of Kamina, Kamina, Democratic Republic of the Congo
| | - Christelle Kalikat Mwavita
- Ministry of Health, National Expanded Program for Immunization, Kinshasa, The Democratic Republic Of Congo
| | - Guillaume Ngoy Mwamba
- Department of Public Health, Faculty of Medicine, University of Kamina, Kamina, Democratic Republic of the Congo
- Ministry of Health, National Expanded Program for Immunization, Kinshasa, The Democratic Republic Of Congo
| | - Aime Cikomola Wa Bene
- Ministry of Health, National Expanded Program for Immunization, Kinshasa, The Democratic Republic Of Congo
| | - Audry Mulumba Wakamba
- Ministry of Health, National Expanded Program for Immunization, Kinshasa, The Democratic Republic Of Congo
| | | | - Michel Kabamba Nzaji
- Department of Public Health, Faculty of Medicine, University of Kamina, Kamina, Democratic Republic of the Congo
- Ministry of Health, National Expanded Program for Immunization, Kinshasa, The Democratic Republic Of Congo
| |
Collapse
|
18
|
Madewell ZJ, Graff NE, Lopez VK, Rodriguez DM, Wong JM, Maniatis P, Medina FA, Muñoz JL, Briggs-Hagen M, Adams LE, Rivera-Amill V, Paz-Bailey G, Major CG. Longitudinal analysis of SARS-CoV-2 IgG antibody durability in Puerto Rico. Sci Rep 2024; 14:30743. [PMID: 39730470 DOI: 10.1038/s41598-024-80465-4] [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: 08/01/2024] [Accepted: 11/19/2024] [Indexed: 12/29/2024] Open
Abstract
Understanding the dynamics of antibody responses following vaccination and SARS-CoV-2 infection is important for informing effective vaccination strategies and other public health interventions. This study investigates SARS-CoV-2 antibody dynamics in a Puerto Rican cohort, analyzing how IgG levels vary by vaccination status and previous infection. We assess waning immunity and the distribution of hybrid immunity with the aim to inform public health strategies and vaccination programs in Puerto Rico and similar settings. We conducted a prospective, longitudinal cohort study to identify SARS-CoV-2 infections and related outcomes in Ponce, Puerto Rico, from June 2020-August 2022. Participants provided self-collected nasal swabs every week and serum every six months for RT-PCR and IgG testing, respectively. IgG reactivity against nucleocapsid (N) antigens, which generally indicate previous infection, and spike (S1) and receptor-binding domain (RBD) antigens, which indicate history of either infection or vaccination, was assessed using the Luminex Corporation xMAP® SARS-CoV-2 Multi-Antigen IgG Assay. Prior infection was defined by positive RT-PCRs, categorized by the predominant circulating SARS-CoV-2 variant at the event time. Demographic information, medical history, and COVID-19 vaccination history were collected through standardized questionnaires. Of 882 participants included in our analysis, 34.0% experienced at least one SARS-CoV-2 infection, with most (78.7%) occurring during the Omicron wave (December 2021 onwards). SARS-CoV-2 antibody prevalence increased over time, reaching 98.4% by the final serum collection, 67.0% attributable to vaccination alone, 1.6% from infection alone, and 31.4% from both. Regardless of prior infection status, RBD and S1 IgG levels gradually declined following two vaccine doses. A third dose boosted these antibody levels and showed a slower decline over time. N-antibody levels peaked during the Omicron surge and waned over time. Vaccination in individuals with prior SARS-CoV-2 infection elicited the highest and most durable antibody responses. N or S1 seropositivity was associated with lower odds of a subsequent positive PCR test during the Omicron period, with N antibodies showing a stronger association. By elucidating the differential decay of RBD and S1 antibodies following vaccination and the complexities of N-antibody response following infection, this study in a Puerto Rican cohort strengthens the foundation for developing targeted interventions and public health strategies.
Collapse
Affiliation(s)
- Zachary J Madewell
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, San Juan, Puerto Rico.
| | - Nathan E Graff
- Coronavirus and Other Respiratory Viruses Division, Centers for Disease Control and Prevention, Atlanta, Georgia
- Eagle Health Analytics, San Antonio, Texas, USA
| | - Velma K Lopez
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, San Juan, Puerto Rico
| | - Dania M Rodriguez
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, San Juan, Puerto Rico
| | - Joshua M Wong
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, San Juan, Puerto Rico
| | - Panagiotis Maniatis
- Division of Bacterial Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Freddy A Medina
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, San Juan, Puerto Rico
| | - Jorge L Muñoz
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, San Juan, Puerto Rico
| | - Melissa Briggs-Hagen
- Coronavirus and Other Respiratory Viruses Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Laura E Adams
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, San Juan, Puerto Rico
| | | | - Gabriela Paz-Bailey
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, San Juan, Puerto Rico
| | - Chelsea G Major
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, San Juan, Puerto Rico
| |
Collapse
|
19
|
Livieratos A, Schiro LE, Gogos C, Akinosoglou K. Durability of Adaptive Immunity in Immunocompetent and Immunocompromised Patients Across Different Respiratory Viruses: RSV, Influenza, and SARS-CoV-2. Vaccines (Basel) 2024; 12:1444. [PMID: 39772104 PMCID: PMC11680120 DOI: 10.3390/vaccines12121444] [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: 12/03/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Research on respiratory virus immunity duration post-vaccination reveals variable outcomes. This study performed a literature review to assess the efficacy and longevity of immune protection post-vaccination against SARS-CoV-2, influenza, and respiratory syncytial virus (RSV), with a focus on immunocompromised populations. Specific objectives included examining humoral and cellular immune responses and exploring the impact of booster doses and hybrid immunity on extending protection. METHODS A literature review was conducted focusing on studies published from January 2014 to November 2024. The search targeted adaptive immunity post-vaccination, natural immunity, and hybrid immunity for SARS-CoV-2, influenza, and RSV. Selection criteria emphasized human populations, adaptive immunity outcomes, and immunocompromised individuals. The PICO framework guided the analysis, culminating in a detailed review of 30 studies. RESULTS SARS-CoV-2 vaccines exhibited robust initial antibody responses, which waned significantly within six months, necessitating frequent boosters. Influenza and RSV vaccines similarly showed declines in immunity, though some influenza vaccines demonstrated moderate durability. Hybrid immunity, arising from combined natural infection and vaccination, provided more resilient and lasting protection than vaccination alone, especially against emerging variants. Immunocompromised individuals consistently exhibited reduced durability in adaptive immune responses across all studied viruses. Challenges include rapid viral mutations, limiting the broad protection of current vaccines. CONCLUSIONS Immune durability varies significantly across virus types and patient populations. Frequent boosters and hybrid immunity are critical to optimizing protection, particularly for vulnerable groups. The findings underscore the need for adaptable vaccination strategies and advancements in vaccine design to counter rapidly mutating respiratory pathogens effectively.
Collapse
Affiliation(s)
| | | | - Charalambos Gogos
- Department of Medicine, University of Patras, 26504 Rio, Greece; (C.G.); (K.A.)
| | - Karolina Akinosoglou
- Department of Medicine, University of Patras, 26504 Rio, Greece; (C.G.); (K.A.)
- Department of Internal Medicine and Infectious Diseases, University General Hospital of Patras, 26504 Rio, Greece
| |
Collapse
|
20
|
Laitin SMD, Baditoiu LM, Laza R, Besliu RS, Stoicescu ER, Gug M, Dumitru CS, Patrascu R. Impact of Vaccination Status on COVID-19 Severity and Pulmonary Involvement. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1919. [PMID: 39768801 PMCID: PMC11678390 DOI: 10.3390/medicina60121919] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025]
Abstract
Background and Objectives: The COVID-19 pandemic has had a significant impact on global health, with serious outcomes, such as lung damage, being major determinants of patient morbidity and mortality. Immunization has been essential in attenuating these outcomes. This study aimed to evaluate the impact of COVID-19 vaccination on disease severity, particularly focusing on pulmonary involvement, among hospitalized patients. Materials and Methods: A retrospective cohort study was conducted at Victor Babes Hospital, Timisoara, involving 3005 patients diagnosed with COVID-19 between December 2020 and March 2022. Patients were stratified into vaccinated and unvaccinated groups. Results: The study found that vaccinated patients had significantly lower rates of severe pulmonary involvement compared to unvaccinated patients. Specifically, only 24.24% of vaccinated patients experienced severe lung involvement, compared to 35.64% in the unvaccinated group (p < 0.001). Vaccinated individuals had shorter hospital stays (8.96 ± 6.40 days vs. 10.70 ± 6.29 days), but this difference was not statistically significant (p = 0.219). Additionally, chronic pulmonary diseases and stroke were less prevalent among vaccinated patients, highlighting the protective effect of vaccination. Conclusions: COVID-19 vaccination significantly reduces the severity of disease, particularly in preventing severe pulmonary involvement, which is a major determinant of patient outcomes. These findings underscore the importance of ongoing vaccination efforts and the need for booster doses to maintain immunity, especially as new variants emerge. The study supports the continued prioritization of vaccination in public health strategies to mitigate the long-term impact of COVID-19.
Collapse
Affiliation(s)
- Sorina Maria Denisa Laitin
- Department XIII, Epidemiology University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (S.M.D.L.); (L.M.B.)
- Clinical Hospital of Infectious Diseases and Pneumology ‘Dr. Victor Babes’ Timisoara, 300310 Timisoara, Romania;
| | - Luminita Mirela Baditoiu
- Department XIII, Epidemiology University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (S.M.D.L.); (L.M.B.)
- Multidisciplinary Research Center on Antimicrobial Resistance, ‘Victor Babes’ University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | - Ruxandra Laza
- Clinical Hospital of Infectious Diseases and Pneumology ‘Dr. Victor Babes’ Timisoara, 300310 Timisoara, Romania;
- Department XIII, Infectious Diseases University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Razvan Sebastian Besliu
- Epidemiology Clinic, ‘Pius Brinzeu’ Emergency Clinical County Hospital Timisoara, Liviu Rebreanu Boulevard No. 156, 300723 Timisoara, Romania;
| | - Emil Robert Stoicescu
- Department XV, Radiology and Medical Imaging University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
- Research Center for Pharmaco-Toxicological Evaluations, ‘Victor Babes’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Field of Applied Engineering Sciences, Specialization Statistical Methods and Techniques in Health and Clinical Research, Faculty of Mechanics, ‘Politehnica’ University Timisoara, Mihai Viteazul Boulevard No. 1, 300222 Timisoara, Romania
| | - Miruna Gug
- Department of Microscopic Morphology, Discipline of Genetics, Doctoral School, ‘Victor Babes’ University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Cristina Stefania Dumitru
- Department of Microscopic Morphology/Histology, ‘Victor Babes’ University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Raul Patrascu
- Department of Functional Sciences, ‘Victor Babes’ University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| |
Collapse
|
21
|
Fischer K, Langley JM, Harrison R, Mubareka S, Nadarajah J, Smieja M, Valiquette L, Cooper C, Powis J, CCS Working Group, McGeer A, Coleman BL. Recent Vaccination Against SARS-CoV-2 Is Associated with Less Severe Disease in Working-Age Adults. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:1501. [PMID: 39595768 PMCID: PMC11594206 DOI: 10.3390/ijerph21111501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Essential workers, including those working in healthcare and education, are at higher risk of exposure to communicable diseases, including SARS-CoV-2. Reducing the rates of infection is important for their personal health and for the ongoing safe operation of essential services. METHODS Data from participants in two prospective cohort studies who tested positive for SARS-CoV-2 in 2020 through 2023 were used to determine whether vaccination against SARS-CoV-2 is associated with the severity of symptoms in working-age adults. RESULTS SARS-CoV-2-positive tests (N = 3757) were reported by 3093 participants (mean: 1.2 per person); 1229 (33%) illnesses did not interfere with regular activities, 1926 (51%) made participants too unwell for regular activities, and 602 (16%) required participant bed rest. Compared with vaccine receipt more than 12 months earlier, receipt within six months of an infection was associated with lower risk ratios for more severe illness (too unwell: 0.69 and bed rest: 0.67) compared with being able to conduct regular activities. More recent vaccination was also associated with lower odds reporting of systemic symptoms (fever, myalgia, arthralgia) and fewer solicited symptoms. CONCLUSION Staying current with COVID-19 vaccinations should continue to be recommended since receiving a recent immunization lessened the severity of illness. Also, as symptoms of COVID-19 are now largely similar to other respiratory viruses, practitioners need to use this evidence to inform diagnostic testing and return-to-work policies.
Collapse
Affiliation(s)
- Kailey Fischer
- Sinai Health, 600 University Ave, Toronto, ON M5G 1X5, Canada; (K.F.)
| | - Joanne M. Langley
- Canadian Center for Vaccinology, Dalhousie University, IWK and Nova Scotia Health, 5850 University Ave, Halifax, NS B3K 6R8, Canada
| | - Robyn Harrison
- Division of Infectious Diseases, University of Alberta, 8440 112 St, Edmonton, AB T5J 3E4, Canada
| | - Samira Mubareka
- Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
| | - Jeya Nadarajah
- Oak Valley Health, 381 Church St, Markham, ON L3P 13P, Canada
| | - Marek Smieja
- St. Joseph’s Healthcare, 50 Charlton Ave East, Hamilton, ON L8N 4A6, Canada
| | - Louis Valiquette
- Centre Hospitalier Universitaire de Sherbrooke, 2500 Bd de l’université, Sherbrooke, QC J1K 2R1, Canada
| | - Curtis Cooper
- Faculty of Medicine, University of Ottawa, 75 Laurier Ave E, Ottawa, ON K1N 6N5, Canada
| | - Jeff Powis
- Michael Garron Hospital, 825 Coxwell Avenue Toronto, ON M4C 3E7, Canada
| | | | - Allison McGeer
- Sinai Health, 600 University Ave, Toronto, ON M5G 1X5, Canada; (K.F.)
- School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, 1 King’s College Cir, Toronto, ON M5S 1A8, Canada
| | - Brenda L. Coleman
- Sinai Health, 600 University Ave, Toronto, ON M5G 1X5, Canada; (K.F.)
- School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| |
Collapse
|
22
|
Fabbri L, Stanel SC. New Evidence for SARS-CoV-2 Vaccine Boosting in Patients With Chronic Lung Diseases. Chest 2024; 166:655-656. [PMID: 39389680 DOI: 10.1016/j.chest.2024.07.155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 10/12/2024] Open
Affiliation(s)
- Laura Fabbri
- National Heart and Lung Institute, Imperial College London, London, England; Department of Respiratory Medicine, Royal Brompton Hospital, London, England.
| | - Stefan Cristian Stanel
- Manchester University NHS Foundation Trust, Manchester, England; University of Manchester, Manchester, England
| |
Collapse
|
23
|
Nguyen HC, Lal KG, Balinsky CA, Hontz RD, Lin J, Beye MJ, Smith L, Pan L, Cheng Y, Fox I, Lizewski SE, Foo HS, Krebs SJ, Sun P, Letizia AG. Informing the Need for a SARS-CoV-2 Booster Based on the Immune Responses Among Young Healthy Adults to Variants Circulating in Late 2023. J Infect Dis 2024; 230:645-656. [PMID: 38718223 DOI: 10.1093/infdis/jiae249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 05/07/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND COVID-19 remains a global public health challenge due to new immune-evasive SARS-CoV-2 variants and heterogeneous immunity. METHODS In this cross-sectional study, we evaluated the adaptive immune responses in US active duty personnel who completed a COVID-19 primary vaccine series and had heterogenous SARS-CoV-2 vaccination and infection histories to 3 previously dominant variants (ancestral, Delta, BA.5) and 3 circulating variants (XBB.1.5, EG.5, and BA.2.86) in late 2023. Analyses were based on the most recent exposure in terms of timing (within or beyond 12 months) and type (vaccine or infection). RESULTS Significant reduction was observed in binding antibodies, neutralization antibodies, memory B cells, and CD8+ T cells against circulating variants when compared with previous variants. The reduction in antibody response was more pronounced in those whose most recent exposure was >12 months from enrollment. In contrast, the CD4+ T-cell response was largely consistent across all tested variants. The type of most recent exposure was not a significant factor in determining the magnitude of current immune responses. CONCLUSIONS Administration of the XBB.1.5-based booster is likely to enhance cross-reactive humoral responses against SARS-CoV-2 circulating lineages. Ongoing surveillance of immune responses to emerging variants is needed for informing vaccine composition and timing.
Collapse
Affiliation(s)
- Huy C Nguyen
- US Naval Medical Research Unit INDO PACIFIC, Science Directorate, Singapore, Singapore
| | - Kerri G Lal
- Walter Reed Army Institute of Research, US Military HIV Research Program, B Cell Biology, Silver Spring
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | - Corey A Balinsky
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
- Naval Medical Research Command, Diagnostics Surveillance Division, Silver Spring
| | - Robert D Hontz
- US Naval Medical Research Unit INDO PACIFIC, Science Directorate, Singapore, Singapore
| | - Jin Lin
- US Naval Medical Research Unit INDO PACIFIC, Science Directorate, Singapore, Singapore
| | - Matthew J Beye
- US Naval Medical Research Unit INDO PACIFIC, Science Directorate, Singapore, Singapore
| | - Lauren Smith
- Walter Reed Army Institute of Research, US Military HIV Research Program, B Cell Biology, Silver Spring
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | - Li Pan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | | | - Isabella Fox
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | - Stephen E Lizewski
- Naval Medical Research Command, Diagnostics Surveillance Division, Silver Spring
| | - Hayley S Foo
- US Naval Medical Research Unit INDO PACIFIC, Science Directorate, Singapore, Singapore
| | - Shelly J Krebs
- Walter Reed Army Institute of Research, US Military HIV Research Program, B Cell Biology, Silver Spring
| | - Peifang Sun
- Naval Medical Research Command, Diagnostics Surveillance Division, Silver Spring
| | - Andrew G Letizia
- US Naval Medical Research Unit INDO PACIFIC, Science Directorate, Singapore, Singapore
| |
Collapse
|
24
|
Elyass J, Desalegn A, Trinh NTH, Orangzeb S, Zidan M, Nordeng H, Lupattelli A. Individual and familial factors associated with mRNA COVID-19 vaccine uptake in pregnancy: A large-scale registry-based linkage study. Vaccine 2024; 42:126171. [PMID: 39079814 DOI: 10.1016/j.vaccine.2024.126171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
The association between maternal COVID-19 vaccination in pregnancy and factors such as high risk for severe COVID-19, pre-existing asthma, prior adverse reproductive history, or paternal COVID-19 vaccination during pregnancy, remains unclear. The aim of this study is two-fold: (i) to describe uptake of COVID-19 vaccine during pregnancy by maternal risk for severe COVID-19 and asthma, and (ii) to comprehensively examine individual and familial factors associated with vaccine uptake during pregnancy in Norway. Based on nation-wide registry-linkage data in Norway, we included 101,659 deliveries with gestational length ≥12 weeks, in 2021-2022. Our outcome measure was uptake of at least one dose of mRNA COVID-19 vaccine during pregnancy, using a narrow (first ever dose) and broad (any dose) definition. We fit univariate and multivariate modified Poisson regression models, clustered by county of residency and adjusted for calendar time, to estimate risk ratios (RR) with 95 % Confidence Intervals (CIs). Gestational uptake of any COVID-19 vaccine dose increased from <1 % before mid Aug-2021, to 38.8 % in the rest of 2021, and 48.9 % in 2022. Only 28.8 % and 33.9 % pregnant individuals with high risk for severe COVID-19 or asthma, respectively, received at least one COVID-19 vaccine dose. Paternal COVID-19 vaccination was strongly associated with greater vaccine uptake by pregnant individuals (adjusted RR: 7.2, 95 % CI: 6.8-7.5). Maternal SARS-CoV-2 infection pre-pregnancy (adjusted RR: 0.31, 95 % CI: 0.26, 0.37), familial and individual migrant status were associated with a considerable decreased likelihood of vaccine uptake in pregnancy. History of miscarriage or pregnancy with congenital anomaly were not associated with vaccine uptake. Despite rising COVID-19 vaccine rates in pregnancy, uptake remained low for high-risk individuals. Paternal vaccination, pre-pregnancy infection, migration status, and maternal citizenship were strongly associated with prenatal vaccine uptake. This knowledge can inform tailoring of future vaccination campaigns.
Collapse
Affiliation(s)
- Jovan Elyass
- PharmacoEpidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway; Université Bordeaux, INSERM, Bordeaux Population Health, Bordeaux, France
| | - Anteneh Desalegn
- PharmacoEpidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Nhung T H Trinh
- PharmacoEpidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Saima Orangzeb
- PharmacoEpidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Mahmoud Zidan
- PharmacoEpidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Hedvig Nordeng
- PharmacoEpidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway; Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| | - Angela Lupattelli
- PharmacoEpidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
25
|
Edwards CT, Karunakaran KA, Garcia E, Beutler N, Gagne M, Golden N, Aoued H, Pellegrini KL, Burnett MR, Honeycutt CC, Lapp SA, Ton T, Lin MC, Metz A, Bombin A, Goff K, Scheuermann SE, Wilkes A, Wood JS, Ehnert S, Weissman S, Curran EH, Roy M, Dessasau E, Paiardini M, Upadhyay AA, Moore I, Maness NJ, Douek DC, Piantadosi A, Andrabi R, Rogers TR, Burton DR, Bosinger SE. Passive infusion of an S2-Stem broadly neutralizing antibody protects against SARS-CoV-2 infection and lower airway inflammation in rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605768. [PMID: 39109178 PMCID: PMC11302620 DOI: 10.1101/2024.07.30.605768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The continued evolution of SARS-CoV-2 variants capable of subverting vaccine and infection-induced immunity suggests the advantage of a broadly protective vaccine against betacoronaviruses (β-CoVs). Recent studies have isolated monoclonal antibodies (mAbs) from SARS-CoV-2 recovered-vaccinated donors capable of neutralizing many variants of SARS-CoV-2 and other β-CoVs. Many of these mAbs target the conserved S2 stem region of the SARS-CoV-2 spike protein, rather the receptor binding domain contained within S1 primarily targeted by current SARS-CoV-2 vaccines. One of these S2-directed mAbs, CC40.8, has demonstrated protective efficacy in small animal models against SARS-CoV-2 challenge. As the next step in the pre-clinical testing of S2-directed antibodies as a strategy to protect from SARS-CoV-2 infection, we evaluated the in vivo efficacy of CC40.8 in a clinically relevant non-human primate model by conducting passive antibody transfer to rhesus macaques (RM) followed by SARS-CoV-2 challenge. CC40.8 mAb was intravenously infused at 10mg/kg, 1mg/kg, or 0.1 mg/kg into groups (n=6) of RM, alongside one group that received a control antibody (PGT121). Viral loads in the lower airway were significantly reduced in animals receiving higher doses of CC40.8. We observed a significant reduction in inflammatory cytokines and macrophages within the lower airway of animals infused with 10mg/kg and 1mg/kg doses of CC40.8. Viral genome sequencing demonstrated a lack of escape mutations in the CC40.8 epitope. Collectively, these data demonstrate the protective efficiency of broadly neutralizing S2-targeting antibodies against SARS-CoV-2 infection within the lower airway while providing critical preclinical work necessary for the development of pan-β-CoV vaccines.
Collapse
Affiliation(s)
- Christopher T. Edwards
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Kirti A. Karunakaran
- Department of Pathology, Microbiology & Immunology, Vanderbilt University, Nashville, TN 37235, USA
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, Minnesota 55356, USA
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Matthew Gagne
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nadia Golden
- Tulane National Primate Research Center, Covington, LA, USA
| | - Hadj Aoued
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Kathryn L. Pellegrini
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Matthew R. Burnett
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Cole Honeycutt
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stacey A. Lapp
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Thang Ton
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Mark C. Lin
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Amanda Metz
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Andrei Bombin
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Kelly Goff
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Amelia Wilkes
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jennifer S. Wood
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Stephanie Ehnert
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Stacey Weissman
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Elizabeth H. Curran
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Melissa Roy
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Evan Dessasau
- Division of Histology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Amit A. Upadhyay
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Ian Moore
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Daniel C. Douek
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anne Piantadosi
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Thomas R. Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139, USA
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
26
|
Le Gars M, Sadoff J, Cárdenas V, Heerwegh D, Tesfaye F, Roey GV, Spicer C, Matias SS, Crayne O, Kamphuis T, Struyf F, Schuitemaker H, Douoguih M. Safety, reactogenicity, and immunogenicity of Ad26.COV2.S as homologous or heterologous COVID-19 booster vaccination: Results of a randomized, double-blind, phase 2 trial. Vaccine 2024; 42:3938-3952. [PMID: 38918103 DOI: 10.1016/j.vaccine.2024.03.079] [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: 12/04/2023] [Revised: 03/29/2024] [Accepted: 03/29/2024] [Indexed: 06/27/2024]
Abstract
COVID-19 vaccine boosters may optimize durability of protection against variants of concern (VOCs). In this randomized, double-blind, phase 2 trial, participants received 3 different dose levels of an Ad26.COV2.S booster (5 × 1010 vp [viral particles], 2.5 × 1010 vp, or 1 × 1010 vp) ≥6 months post-primary vaccination with either single-dose Ad26.COV2.S (homologous boost; n = 774) or 2-dose BNT162b2 (heterologous boost; n = 758). Primary endpoints were noninferiority of neutralizing antibody responses at Day 15 post-boost versus Day 29 post-primary vaccination. Secondary endpoints included reactogenicity/safety and neutralizing antibody responses to VOCs. All primary endpoints passed prespecified hierarchical noninferiority criteria by Day 15 post-boost. Geometric mean increases in neutralizing antibody titers against the D614G reference strain ranged from 5.5 to 6.8 at Day 15 for homologous boosting and 12.6 to 22.0 for heterologous boosting. For VOCs, heterologous boosting elicited higher neutralizing antibody responses than homologous boosting. Neutralizing antibody responses were dose-dependent and durable for ≥6 months post-boost. More solicited systemic adverse events occurred following heterologous versus homologous boosting. Trial Registration:ClinicalTrials.gov Identifier: NCT04999111.
Collapse
Affiliation(s)
- Mathieu Le Gars
- Janssen Vaccines & Prevention, Newtonweg 1, 2333 CN Leiden, The Netherlands.
| | - Jerald Sadoff
- Johnson & Johnson, 1 Johnson & Johnson Plaza, New Brunswick, NJ, USA.
| | - Vicky Cárdenas
- Janssen Research & Development, 1400 McKean Rd., Spring House, PA, USA.
| | - Dirk Heerwegh
- Janssen Research & Development, Turnhoutseweg 30, Beerse, Belgium.
| | - Fisseha Tesfaye
- Janssen Research & Development, 1000 U.S. Route 202 South, Raritan, NJ, USA.
| | - Griet Van Roey
- Janssen Vaccines & Prevention, Newtonweg 1, 2333 CN Leiden, The Netherlands.
| | - Colleen Spicer
- Janssen Research & Development, 1000 U.S. Route 202 South, Raritan, NJ, USA.
| | | | - Olivia Crayne
- Janssen Research & Development, 1000 U.S. Route 202 South, Raritan, NJ, USA.
| | - Tobias Kamphuis
- Janssen Vaccines & Prevention, Newtonweg 1, 2333 CN Leiden, The Netherlands.
| | - Frank Struyf
- Janssen Research & Development, Turnhoutseweg 30, Beerse, Belgium.
| | | | - Macaya Douoguih
- Janssen Vaccines & Prevention, Newtonweg 1, 2333 CN Leiden, The Netherlands.
| |
Collapse
|
27
|
Hofmeyer KA, Ventura CL, Armstrong KL, Houchens CR, Patel S, Disbrow GL, Johnson RA. Project NextGen: Developing the Next Generation of COVID-19 Vaccines and Therapeutics to Respond to the Present and Prepare for the Future. Clin Infect Dis 2024; 79:115-121. [PMID: 38356144 PMCID: PMC11259220 DOI: 10.1093/cid/ciae073] [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: 11/01/2023] [Revised: 01/03/2024] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) epidemiology and product landscapes have changed considerably since onset of the pandemic. Safe and effective vaccines and therapeutics are available, but the continual emergence of severe acute respiratory syndrome coronavirus 2 variants introduce limitations in our ability to prevent and treat disease. Project NextGen is a collaboration between the Biomedical Advanced Research and Development Authority, part of the Administration for Strategic Preparedness and Response, and the National Institute of Allergy and Infectious Diseases, part of the National Institutes of Health, that is leveraging public-private partnerships to address gaps in the nation's COVID-19 vaccine and therapeutic capabilities. Targeted investments will advance promising next-generation candidates through the most difficult phases of clinical development to encourage further private sector interest for later stage development and commercial availability. New commercial vaccines and therapeutics that are more durable and effective across variants will improve our fight against COVID-19 and transform our response to future threats.
Collapse
Affiliation(s)
- Kimberly A Hofmeyer
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Christy L Ventura
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Kimberly L Armstrong
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Christopher R Houchens
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Sandeep Patel
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Gary L Disbrow
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Robert A Johnson
- Biomedical Advanced Research and Development Authority, Administration for Strategic Preparedness and Response, U.S. Department of Health and Human Services, Washington, DC, USA
| |
Collapse
|
28
|
Tan MW, Anelone AJN, Tay AT, Tan RY, Zeng K, Tan KB, Clapham HE. Differences in virus and immune dynamics for SARS-CoV-2 Delta and Omicron infections by age and vaccination histories. BMC Infect Dis 2024; 24:654. [PMID: 38951848 PMCID: PMC11218222 DOI: 10.1186/s12879-024-09572-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/27/2024] [Indexed: 07/03/2024] Open
Abstract
Vaccination against COVID-19 was integral to controlling the pandemic that persisted with the continuous emergence of SARS-CoV-2 variants. Using a mathematical model describing SARS-CoV-2 within-host infection dynamics, we estimate differences in virus and immunity due to factors of infecting variant, age, and vaccination history (vaccination brand, number of doses and time since vaccination). We fit our model in a Bayesian framework to upper respiratory tract viral load measurements obtained from cases of Delta and Omicron infections in Singapore, of whom the majority only had one nasopharyngeal swab measurement. With this dataset, we are able to recreate similar trends in URT virus dynamics observed in past within-host modelling studies fitted to longitudinal patient data.We found that Omicron had higher R0,within values than Delta, indicating greater initial cell-to-cell spread of infection within the host. Moreover, heterogeneities in infection dynamics across patient subgroups could be recreated by fitting immunity-related parameters as vaccination history-specific, with or without age modification. Our model results are consistent with the notion of immunosenescence in SARS-CoV-2 infection in elderly individuals, and the issue of waning immunity with increased time since last vaccination. Lastly, vaccination was not found to subdue virus dynamics in Omicron infections as well as it had for Delta infections.This study provides insight into the influence of vaccine-elicited immunity on SARS-CoV-2 within-host dynamics, and the interplay between age and vaccination history. Furthermore, it demonstrates the need to disentangle host factors and changes in pathogen to discern factors influencing virus dynamics. Finally, this work demonstrates a way forward in the study of within-host virus dynamics, by use of viral load datasets including a large number of patients without repeated measurements.
Collapse
Affiliation(s)
- Maxine W Tan
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore.
| | - Anet J N Anelone
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | | | | | - Kangwei Zeng
- Ministry of Health, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Kelvin Bryan Tan
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
- Ministry of Health, Singapore, Singapore
- Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore
| | - Hannah Eleanor Clapham
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| |
Collapse
|
29
|
Claus J, ten Doesschate T, Taks E, Debisarun PA, Smits G, van Binnendijk R, van der Klis F, Verhagen LM, de Jonge MI, Bonten MJM, Netea MG, van de Wijgert JHHM. Determinants of Systemic SARS-CoV-2-Specific Antibody Responses to Infection and to Vaccination: A Secondary Analysis of Randomised Controlled Trial Data. Vaccines (Basel) 2024; 12:691. [PMID: 38932420 PMCID: PMC11209274 DOI: 10.3390/vaccines12060691] [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: 05/17/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
SARS-CoV-2 infections elicit antibodies against the viral spike (S) and nucleocapsid (N) proteins; COVID-19 vaccines against the S-protein only. The BCG-Corona trial, initiated in March 2020 in SARS-CoV-2-naïve Dutch healthcare workers, captured several epidemic peaks and the introduction of COVID-19 vaccines during the one-year follow-up. We assessed determinants of systemic anti-S1 and anti-N immunoglobulin type G (IgG) responses using trial data. Participants were randomised to BCG or placebo vaccination, reported daily symptoms, SARS-CoV-2 test results, and COVID-19 vaccinations, and donated blood for SARS-CoV-2 serology at two time points. In the 970 participants, anti-S1 geometric mean antibody concentrations (GMCs) were much higher than anti-N GMCs. Anti-S1 GMCs significantly increased with increasing number of immune events (SARS-CoV-2 infection or COVID-19 vaccination): 104.7 international units (IU)/mL, 955.0 IU/mL, and 2290.9 IU/mL for one, two, and three immune events, respectively (p < 0.001). In adjusted multivariable linear regression models, anti-S1 and anti-N log10 concentrations were significantly associated with infection severity, and anti-S1 log10 concentration with COVID-19 vaccine type/dose. In univariable models, anti-N log10 concentration was also significantly associated with acute infection duration, and severity and duration of individual symptoms. Antibody concentrations were not associated with long COVID or long-term loss of smell/taste.
Collapse
Affiliation(s)
- Juana Claus
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (J.C.); (T.t.D.); (J.H.H.M.v.d.W.)
| | - Thijs ten Doesschate
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (J.C.); (T.t.D.); (J.H.H.M.v.d.W.)
- Department of Internal Medicine, Jeroen Bosch Ziekenhuis, 5223 GZ Hertogenbosch, The Netherlands
| | - Esther Taks
- Department of Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (E.T.); (P.A.D.); (M.G.N.)
| | - Priya A. Debisarun
- Department of Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (E.T.); (P.A.D.); (M.G.N.)
| | - Gaby Smits
- National Institute of Public Health and the Environment, 3720 BA Bilthoven, The Netherlands; (G.S.); (R.v.B.); (F.v.d.K.)
| | - Rob van Binnendijk
- National Institute of Public Health and the Environment, 3720 BA Bilthoven, The Netherlands; (G.S.); (R.v.B.); (F.v.d.K.)
| | - Fiona van der Klis
- National Institute of Public Health and the Environment, 3720 BA Bilthoven, The Netherlands; (G.S.); (R.v.B.); (F.v.d.K.)
| | - Lilly M. Verhagen
- Department of Paediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Marien I. de Jonge
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Marc J. M. Bonten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (J.C.); (T.t.D.); (J.H.H.M.v.d.W.)
| | - Mihai G. Netea
- Department of Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (E.T.); (P.A.D.); (M.G.N.)
- Department for Genomics & Immunoregulation, Life and Medical Sciences Institute, University of Bonn, 53113 Bonn, Germany
| | - Janneke H. H. M. van de Wijgert
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (J.C.); (T.t.D.); (J.H.H.M.v.d.W.)
| |
Collapse
|
30
|
Hamzavi SS, Bahrololoom R, Saeb S, Marandi NH, Hosseini M, Hesam Abadi AK, Jamalidoust M. Humoral immune response and safety of Sars-Cov-2 vaccine in people with multiple sclerosis. BMC Immunol 2024; 25:35. [PMID: 38898409 PMCID: PMC11186195 DOI: 10.1186/s12865-024-00628-w] [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: 02/19/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND For the past three years, the pandemic has had a major effect on global public health, mainly on those with underlying medical conditions, such as people living with Multiple Sclerosis. Vaccination among this group is of great importance, and the long-term impacts of vaccination and its safety on the health of these patients will continue to be revealed. Therefore, risks related to vaccination and immune response need to be assessed. The objective here was to characterize the immune response, short-term safety, and the effects of multiple variables on these factors after COVID-19 vaccination (mainly Sinopharm) among people with Multiple Sclerosis. We assessed the short-term safety and humoral SARS-COV-2 anti-RBD IgG response using a data collection form and Immunoassay, respectively. RESULTS No severe adverse events or MS relapse was observed. Myalgia/body pain (26.7%), low-grade fever (22.2%), and mild headache (15.6%) were the most common adverse events. The use and type of vaccine influenced the frequency of side effects with a p-value < 0.0001. Regarding immune response, patients on rituximab and fingolimod had a lower antibody titer compared to other medications. With a significant difference, hybrid immunity (p-value: 0.047) and type of DMTs (p-value: 0.017) affected the humoral response. CONCLUSION There is a low incidence of serious adverse effects, MS worsening or relapse after COVID-19 vaccination, and mainly, side effects are similar to that of the general population. It appears that treatment with various disease-modifying therapies does not induce or worsen the post-vaccination side effects, although some, including Rituximab and fingolimod, may affect the immunity induced after vaccination.
Collapse
Affiliation(s)
- Seyedeh Sadigheh Hamzavi
- Department of Pediatrics, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
- Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, 71936-13311, Iran
| | - Rosemina Bahrololoom
- Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, 71936-13311, Iran.
| | - Sepideh Saeb
- Department of Virology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Nahid Heydari Marandi
- Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, 71936-13311, Iran
| | - Marzieh Hosseini
- Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, 71936-13311, Iran
| | | | - Marzieh Jamalidoust
- Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, 71936-13311, Iran.
| |
Collapse
|
31
|
De Bouver C, Bouziotis J, Wijtvliet VPWM, Ariën KK, Mariën J, Heyndrickx L, Couttenye MM, de Fijter HJW, Mestrez F, Treille S, Mat O, Collart F, Allard SD, Vingerhoets L, Moons P, Abramowicz D, De Winter BY, Pipeleers L, Wissing KM, Ledeganck KJ. Humoral immunity to SARS-CoV-2 in kidney transplant recipients and dialysis patients: IgA and IgG patterns unraveled after SARS-CoV-2 infection and vaccination. Virol J 2024; 21:138. [PMID: 38872127 PMCID: PMC11170792 DOI: 10.1186/s12985-024-02410-1] [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: 03/20/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Infection with SARS-CoV-2 in high-risk groups such as kidney transplant and dialysis patients is shown to be associated with a more serious course of the disease. Four years after the start of the COVID-19 pandemic, crucial knowledge on the immune responses in these patient groups is still lacking. Therefore, this study aimed at investigating the humoral immune response after a SARS-CoV-2 infection compared to vaccination as well as the evolution of immunoglobulins over time. METHODS Kidney transplant recipients, patients on haemodialysis or on peritoneal dialysis and healthy controls were included in this longitudinal multicenter study. SARS-CoV-2 anti-RBD, anti-NP and anti-S1S2 immunoglobulin G (IgG) and A (IgA) as well as the neutralizing antibody capacity were measured. RESULTS Kidney transplant recipients had a significantly better humoral response to SARS-CoV-2 after infection (86.4%) than after a two-dose mRNA vaccination (55.8%) while seroconversion was comparable in patients on haemodialysis after infection (95.8%) versus vaccination (89.4%). In individuals without prior COVID-19, the IgG levels after vaccination were significantly lower in kidney transplant recipients when compared to all other groups. However, the IgA titres remained the highest in this patient group at each time point, both after infection and vaccination. A history COVID-19 was associated with higher antibody levels after double-dose vaccination in all patient categories and, while decreasing, titres remained high six months after double-dose vaccination. CONCLUSION Kidney transplant recipients had a more robust humoral response to SARS-CoV-2 following infection compared to a two-dose mRNA vaccination, while patients on haemodialysis exhibited comparable seroconversion rates. Notably, individuals with prior COVID-19 exhibited higher IgG levels in response to vaccination. Hybrid immunity is thus the best possible defence against severe COVID-19 disease and seems also to hold up for these populations. Next, it is not clear whether the higher IgA levels in the kidney transplant recipients is beneficial for neutralizing SARS-CoV-2 or if it is a sign of disease severity.
Collapse
Affiliation(s)
- Caroline De Bouver
- Laboratory of Experimental Medicine and Pediatrics and member of the Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jason Bouziotis
- Clinical Trial Center (CTC), CRC Antwerp, Antwerp University Hospital, Edegem, Belgium
| | - Veerle P W M Wijtvliet
- Laboratory of Experimental Medicine and Pediatrics and member of the Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Department of Nephrology and Hypertension, Antwerp University Hospital, Edegem, Belgium
| | - Kevin K Ariën
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Joachim Mariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Leo Heyndrickx
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Marie M Couttenye
- Laboratory of Experimental Medicine and Pediatrics and member of the Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Department of Nephrology and Hypertension, Antwerp University Hospital, Edegem, Belgium
| | - Hans J W de Fijter
- Laboratory of Experimental Medicine and Pediatrics and member of the Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Department of Nephrology and Hypertension, Antwerp University Hospital, Edegem, Belgium
| | - Fabienne Mestrez
- Department of Nephrology-Dialysis, University Hospital (CHU) Ambroise Paré, Mons, Belgium
| | - Serge Treille
- Department of Nephrology, Centre Hospitalier Universitaire Charleroi, Charleroi, Belgium
| | - Olivier Mat
- Department of Nephrology, Hospital Centre EpiCURA, Ath, Belgium
| | - Frederic Collart
- Department of Nephrology, Hospital Universitaire Brugmann, Brussels, Belgium
| | - Sabine D Allard
- Department of Internal Medicine and Infectious Diseases, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | | | - Pieter Moons
- Biobank Antwerp, Antwerp University Hospital, Edegem, Belgium
| | - Daniel Abramowicz
- Laboratory of Experimental Medicine and Pediatrics and member of the Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Department of Nephrology and Hypertension, Antwerp University Hospital, Edegem, Belgium
| | - Benedicte Y De Winter
- Laboratory of Experimental Medicine and Pediatrics and member of the Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Lissa Pipeleers
- Department of Nephrology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karl Martin Wissing
- Department of Nephrology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kristien J Ledeganck
- Laboratory of Experimental Medicine and Pediatrics and member of the Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
32
|
Ofori SK, Dankwa EA, Estrada EH, Hua X, Kimani TN, Wade CG, Buckee CO, Murray MB, Hedt-Gauthier BL. COVID-19 vaccination strategies in Africa: A scoping review of the use of mathematical models to inform policy. Trop Med Int Health 2024; 29:466-476. [PMID: 38740040 DOI: 10.1111/tmi.13994] [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] [Indexed: 05/16/2024]
Abstract
OBJECTIVE Mathematical models are vital tools to understand transmission dynamics and assess the impact of interventions to mitigate COVID-19. However, historically, their use in Africa has been limited. In this scoping review, we assess how mathematical models were used to study COVID-19 vaccination to potentially inform pandemic planning and response in Africa. METHODS We searched six electronic databases: MEDLINE, Embase, Web of Science, Global Health, MathSciNet and Africa-Wide NiPAD, using keywords to identify articles focused on the use of mathematical modelling studies of COVID-19 vaccination in Africa that were published as of October 2022. We extracted the details on the country, author affiliation, characteristics of models, policy intent and heterogeneity factors. We assessed quality using 21-point scale criteria on model characteristics and content of the studies. RESULTS The literature search yielded 462 articles, of which 32 were included based on the eligibility criteria. Nineteen (59%) studies had a first author affiliated with an African country. Of the 32 included studies, 30 (94%) were compartmental models. By country, most studies were about or included South Africa (n = 12, 37%), followed by Morocco (n = 6, 19%) and Ethiopia (n = 5, 16%). Most studies (n = 19, 59%) assessed the impact of increasing vaccination coverage on COVID-19 burden. Half (n = 16, 50%) had policy intent: prioritising or selecting interventions, pandemic planning and response, vaccine distribution and optimisation strategies and understanding transmission dynamics of COVID-19. Fourteen studies (44%) were of medium quality and eight (25%) were of high quality. CONCLUSIONS While decision-makers could draw vital insights from the evidence generated from mathematical modelling to inform policy, we found that there was limited use of such models exploring vaccination impacts for COVID-19 in Africa. The disparity can be addressed by scaling up mathematical modelling training, increasing collaborative opportunities between modellers and policymakers, and increasing access to funding.
Collapse
Affiliation(s)
- Sylvia K Ofori
- Center for Communicable Disease Dynamics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Emmanuelle A Dankwa
- Center for Communicable Disease Dynamics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Eve Hiyori Estrada
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Xinyi Hua
- Department of Biostatistics, Epidemiology and Environmental Health Sciences, Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro, Georgia, USA
| | - Teresia N Kimani
- KAVI-Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
- Center for Epidemiological Modelling and Analysis, University of Nairobi, Nairobi, Kenya
- Paul G Allen School for Global Animal Health, Washington State University, Pullman, Washington, USA
- Department of Health Services, Kiambu County, Ministry of Health Kenya, Kiambu County, Kenya
| | - Carrie G Wade
- Countway Library, Harvard School of Medicine, Boston, Massachusetts, USA
| | - Caroline O Buckee
- Center for Communicable Disease Dynamics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Megan B Murray
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Bethany L Hedt-Gauthier
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Pasev M, Trifonova A, Velichkov A, Terzieva V. Duration of antibody response to the receptor binding domain of SARS-CoV-2 in infected or vaccinated individuals - A one year retrospective cohort study. Int Immunopharmacol 2024; 133:112084. [PMID: 38621337 DOI: 10.1016/j.intimp.2024.112084] [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: 11/21/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024]
Abstract
The 2019 coronavirus (COVID-19) pandemic raised many scientific and medical questions. Of interest are the duration and effectiveness of the humoral immune response, especially since part of the pandemic occurred in the presence of anti-SARS-CoV-2 vaccines. We retrospectively studied 564 serum samples from 393 post-infected and vaccinated individuals to investigate the longevity and magnitude of the anti-spike IgG response. Our results showed that SARS-CoV-2 anti-spike IgG antibodies are retained for nine-twelve months, in both groups. In the vaccinated group we found higher IgG levels, but with a steeper decrease in titer over the study period. The recovered group's antibody levels correlated well with the national infection trendline for 2021. Both groups showed different, but distinct neutralizing capabilities towards RBD. The anti-Spike IgG response was sustained and efficient, independently of the triggering event, infection or vaccination, with the adaptive capacity against new viral variants being more valuable after infection.
Collapse
Affiliation(s)
- Martin Pasev
- Department of Clinical Microbiology and Virology, University Hospital Lozenetz, Sofia University "Sv. Kliment Ohridski", Sofia, Bulgaria
| | - Angelina Trifonova
- Department of Clinical Microbiology and Virology, University Hospital Lozenetz, Sofia University "Sv. Kliment Ohridski", Sofia, Bulgaria
| | - Andrey Velichkov
- Laboratory of Reproductive OMICs Technologies, Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Velislava Terzieva
- Department of Clinical Microbiology and Virology, University Hospital Lozenetz, Sofia University "Sv. Kliment Ohridski", Sofia, Bulgaria; Laboratory of Reproductive OMICs Technologies, Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria.
| |
Collapse
|
34
|
Hu WC, Chiu SK, Yang YF, Singh S. COVID-19 Vaccination Reporting and Adverse Event Analysis in Taiwan. Vaccines (Basel) 2024; 12:591. [PMID: 38932320 PMCID: PMC11209125 DOI: 10.3390/vaccines12060591] [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: 04/05/2024] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
The COVID-19 pandemic necessitated an urgent global response in vaccine deployment, achieving over 70.6% global vaccination coverage with at least one dose. This study focuses on Taiwan's vaccine administration and adverse event reporting, set against a global backdrop. Using data from Taiwan's Vaccine Adverse Event Reporting System (VAERS) and global vaccination data, this study investigates vaccine safety and the public health implications of vaccination strategies from local and global perspectives. Taiwan's proactive approach, resulting in high vaccination rates, provides a case study for the monitoring and management of vaccine-related adverse events. This study offers insights into the safety profiles of various COVID-19 vaccines and further explores the implications of adverse event reporting rates for vaccine policy and public health strategies. The comparative analysis reveals that, while vaccination has been effective in controlling the virus's spread, safety monitoring remains critical for maintaining public trust. It underscores the necessity of enhanced surveillance and the importance of transparent and tailored risk communication to support informed public health decisions. The findings aim to contribute to the global dialogue on vaccine safety, equitable distribution, evidence-based policy-making, and development of mitigation measures with consideration of local demographics in the ongoing fight against COVID-19.
Collapse
Affiliation(s)
- Wan-Chung Hu
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan;
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
- Department of Biotechnology, Ming Chuan University, Taoyuan 333, Taiwan
| | - Sheng-Kang Chiu
- Division of Infection Diseases, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan;
- School of Medicine, Tzu Chi University, Hualien 970, Taiwan
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Ying-Fei Yang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Sher Singh
- Department of Life Science, School of Life Science, College of Science, National Taiwan Normal University, Taipei 11677, Taiwan
| |
Collapse
|
35
|
Haq MA, Roy AK, Ahmed R, Kuddusi RU, Sinha M, Hossain MS, Vandenent M, Islam MZ, Zaman RU, Kibria MG, Razzaque A, Raqib R, Sarker P. Antibody longevity and waning following COVID-19 vaccination in a 1-year longitudinal cohort in Bangladesh. Sci Rep 2024; 14:11467. [PMID: 38769324 PMCID: PMC11106241 DOI: 10.1038/s41598-024-61922-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024] Open
Abstract
COVID-19 vaccines have been effective in preventing severe illness, hospitalization and death, however, the effectiveness diminishes with time. Here, we evaluated the longevity of antibodies generated by COIVD-19 vaccines and the risk of (re)infection in Bangladeshi population. Adults receiving two doses of AstraZeneca, Pfizer, Moderna or Sinopharm vaccines were enrolled at 2-4 weeks after second dosing and followed-up at 4-monthly interval for 1 year. Data on COVID-like symptoms, confirmed COVID-19 infection, co-morbidities, and receipt of booster dose were collected; blood was collected for measuring spike (S)- and nucleocapsid (N)-specific antibodies. S-specific antibody titers reduced by ~ 50% at 1st follow-up visit and continued to decline unless re-stimulated by booster vaccine dose or (re)infection. Individuals infected between follow-up visits showed significantly lower S-antibody titers at preceding visits compared to the uninfected individuals. Pre-enrolment infection between primary vaccination dosing exhibited 60% and 50% protection against reinfection at 5 and 9 months, respectively. mRNA vaccines provided highest odds of protection from (re)infection up to 5 months (Odds Ratio (OR) = 0.08), however, protection persisted for 9 months in AstraZeneca vaccine recipients (OR = 0.06). In conclusion, vaccine-mediated protection from (re)infection is partially linked to elevated levels of S-specific antibodies. AstraZeneca vaccine provided the longest protection.
Collapse
Affiliation(s)
- Md Ahsanul Haq
- Immunobiology, Nutrition and Toxicology Laboratory, Nutrition Research Division, International Center for Diarrhoeal Disease Research, Bangladesh (icddr, b), Dhaka, 1212, Bangladesh
| | - Anjan Kumar Roy
- Immunobiology, Nutrition and Toxicology Laboratory, Nutrition Research Division, International Center for Diarrhoeal Disease Research, Bangladesh (icddr, b), Dhaka, 1212, Bangladesh
| | - Razu Ahmed
- Immunobiology, Nutrition and Toxicology Laboratory, Nutrition Research Division, International Center for Diarrhoeal Disease Research, Bangladesh (icddr, b), Dhaka, 1212, Bangladesh
| | - Rakib Ullah Kuddusi
- Immunobiology, Nutrition and Toxicology Laboratory, Nutrition Research Division, International Center for Diarrhoeal Disease Research, Bangladesh (icddr, b), Dhaka, 1212, Bangladesh
| | - Monika Sinha
- Immunobiology, Nutrition and Toxicology Laboratory, Nutrition Research Division, International Center for Diarrhoeal Disease Research, Bangladesh (icddr, b), Dhaka, 1212, Bangladesh
| | - Md Shamim Hossain
- Immunobiology, Nutrition and Toxicology Laboratory, Nutrition Research Division, International Center for Diarrhoeal Disease Research, Bangladesh (icddr, b), Dhaka, 1212, Bangladesh
| | | | | | | | - Md Golam Kibria
- Sheikh Russel Gastroliver Institute and Hospital, Dhaka, 1212, Bangladesh
| | - Abdur Razzaque
- Immunobiology, Nutrition and Toxicology Laboratory, Nutrition Research Division, International Center for Diarrhoeal Disease Research, Bangladesh (icddr, b), Dhaka, 1212, Bangladesh
| | - Rubhana Raqib
- Immunobiology, Nutrition and Toxicology Laboratory, Nutrition Research Division, International Center for Diarrhoeal Disease Research, Bangladesh (icddr, b), Dhaka, 1212, Bangladesh
| | - Protim Sarker
- Immunobiology, Nutrition and Toxicology Laboratory, Nutrition Research Division, International Center for Diarrhoeal Disease Research, Bangladesh (icddr, b), Dhaka, 1212, Bangladesh.
| |
Collapse
|
36
|
Grayo S, Sagno H, Diassy O, Zogbelemou JB, Kondabo SJ, Houndekon M, Dellagi K, Vigan-Womas I, Rourou S, Hamouda WB, Benabdessalem C, Ahmed MB, Tordo N. Snapshot of Anti-SARS-CoV-2 IgG Antibodies in COVID-19 Recovered Patients in Guinea. J Clin Med 2024; 13:2965. [PMID: 38792506 PMCID: PMC11122401 DOI: 10.3390/jcm13102965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
Background: Because the regular vaccine campaign started in Guinea one year after the COVID-19 index case, the profile of naturally acquired immunity following primary SARS-CoV-2 infection needs to be deepened. Methods: Blood samples were collected once from 200 patients (90% of African extraction) who were recovered from COVID-19 for at least ~2.4 months (72 days), and their sera were tested for IgG antibodies to SARS-CoV-2 using an in-house ELISA assay against the Receptor Binding Domain (RBD) of the SARS-CoV-2 spike1 protein (RBD/S1-IH kit). Results: Results revealed that 73% of sera (146/200) were positive for IgG to SARS-CoV-2 with an Optical Density (OD) ranging from 0.13 to 1.19 and a median value of 0.56 (IC95: 0.51-0.61). The median OD value at 3 months (1.040) suddenly decreased thereafter and remained stable around OD 0.5 until 15 months post-infection. The OD median value was slightly higher in males compared to females (0.62 vs. 0.49), but the difference was not statistically significant (p-value: 0.073). In contrast, the OD median value was significantly higher among the 60-100 age group (0.87) compared to other groups, with a noteworthy odds ratio compared to the 0-20 age group (OR: 9.69, p-value: 0.044*). Results from the RBD/S1-IH ELISA kit demonstrated superior concordance with the whole spike1 protein ELISA commercial kit compared to a nucleoprotein ELISA commercial kit. Furthermore, anti-spike1 protein ELISAs (whole spike1 and RBD/S1) revealed higher seropositivity rates. Conclusions: These findings underscore the necessity for additional insights into naturally acquired immunity against COVID-19 and emphasize the relevance of specific ELISA kits for accurate seropositivity rates.
Collapse
Affiliation(s)
- Solène Grayo
- Institut Pasteur de Guinée, Conakry BP 4416, Guinea; (H.S.); (N.T.)
| | - Houlou Sagno
- Institut Pasteur de Guinée, Conakry BP 4416, Guinea; (H.S.); (N.T.)
| | - Oumar Diassy
- Agence Nationale de Sécurité Sanitaire, Conakry BP 797, Guinea;
| | | | | | - Marilyn Houndekon
- Centre Médico-Social de L’ambassade de France, Conakry BP 295, Guinea; (J.-B.Z.); (M.H.)
| | - Koussay Dellagi
- Direction Internationale, Institut Pasteur, 75724 Paris, France;
| | | | - Samia Rourou
- Institut Pasteur de Tunis, Tunis BP 74-1002, Tunisia; (S.R.); : (C.B.); (M.B.A.)
| | - Wafa Ben Hamouda
- Institut Pasteur de Tunis, Tunis BP 74-1002, Tunisia; (S.R.); : (C.B.); (M.B.A.)
| | | | - Melika Ben Ahmed
- Institut Pasteur de Tunis, Tunis BP 74-1002, Tunisia; (S.R.); : (C.B.); (M.B.A.)
| | - Noël Tordo
- Institut Pasteur de Guinée, Conakry BP 4416, Guinea; (H.S.); (N.T.)
| |
Collapse
|
37
|
Williamson GL, Bachelder EM, Ainslie KM. Clinical and Preclinical Methods of Heat-Stabilization of Human Vaccines. Mol Pharm 2024; 21:1015-1026. [PMID: 38288698 PMCID: PMC11607726 DOI: 10.1021/acs.molpharmaceut.3c00844] [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] [Indexed: 03/05/2024]
Abstract
Vaccines have historically faced challenges regarding stability, especially in regions lacking a robust cold chain infrastructure. This review delves into established and emergent techniques to improve the thermostability of vaccines. We discuss the widely practiced lyophilization method, effectively transforming liquid vaccine formulations into a solid powdered state, enhancing storage and transportation ability. However, potential protein denaturation during lyophilization necessitates alternative stabilization methods. Cryoprotectants, namely, starch and sugar molecules, have shown promise in protecting vaccine antigens and adjuvants from denaturation and augmenting the stability of biologics during freeze-drying. Biomineralization, a less studied yet innovative approach, utilizes inorganic or organic-inorganic hybrids to encapsulate biological components of vaccines with a particular emphasis on metal-organic coordination polymers. Encapsulation in organic matrices to form particles or microneedles have also been studied in the context of vaccine thermostability, showing some ability to store outside the cold-chain. Unfortunately, few of these techniques have advanced to clinical trials that evaluate differences in storage conditions. Nonetheless, early trials suggest that alternative storage techniques are viable and emphasize the need for more comprehensive studies. This review underscores the pressing need for heat-stable vaccines, especially in light of the increasing global distribution challenges. Combining traditional methods with novel approaches holds promise for the future adaptability of vaccine distribution and use.
Collapse
Affiliation(s)
- Grace L. Williamson
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA
- Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, School of Medicine, UNC, Chapel Hill, NC, USA
| |
Collapse
|
38
|
Aboshi M, Matsuda K, Kawakami D, Kono K, Kazami Y, Sekida T, Komori M, Morey AL, Suga S, Smith JF, Fukuhara T, Iwatani Y, Yamamoto T, Sato N, Akahata W. Safety and immunogenicity of VLPCOV-02, a SARS-CoV-2 self-amplifying RNA vaccine with a modified base, 5-methylcytosine. iScience 2024; 27:108964. [PMID: 38352232 PMCID: PMC10863314 DOI: 10.1016/j.isci.2024.108964] [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: 09/15/2023] [Revised: 11/15/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024] Open
Abstract
Continuing emergence of variants of concern resulting in reduced SARS-CoV-2 vaccine efficacy necessitates additional prevention strategies. The structure of VLPCOV-01, a lipid nanoparticle-encapsulated, self-amplifying RNA COVID-19 vaccine with a comparable immune response to BNT162b2, was revised by incorporating a modified base, 5-methylcytosine, to reduce reactogenicity, and an updated receptor-binding domain derived from the Brazil (gamma) variant. Interim analyses of a phase 1 dose-escalation booster vaccination study with the resulting construct, VLPCOV-02, in healthy, previously vaccinated Japanese individuals (N = 96) are reported (jRCT2051230005). A dose-related increase in solicited local and systemic adverse events was observed, which were generally rated mild or moderate. The most commonly occurring events were tenderness, pain, fatigue, and myalgia. Serum SARS-CoV-2 immunoglobulin titers increased during the 4 weeks post-immunization. VLPCOV-02 demonstrated a favorable safety profile compared with VLPCOV-01, with reduced adverse events and fewer fever events at an equivalent dose. These findings support further study of VLPCOV-02.
Collapse
Affiliation(s)
- Masayuki Aboshi
- VLP Therapeutics Japan, Inc., 1-16-4 Nishi-Shinbashi, Minato-ku, Tokyo 105-0003, Japan
| | | | - Daisuke Kawakami
- VLP Therapeutics Japan, Inc., 1-16-4 Nishi-Shinbashi, Minato-ku, Tokyo 105-0003, Japan
| | - Kaoru Kono
- VLP Therapeutics Japan, Inc., 1-16-4 Nishi-Shinbashi, Minato-ku, Tokyo 105-0003, Japan
| | - Yoko Kazami
- VLP Therapeutics Japan, Inc., 1-16-4 Nishi-Shinbashi, Minato-ku, Tokyo 105-0003, Japan
| | - Takashi Sekida
- VLP Therapeutics Japan, Inc., 1-16-4 Nishi-Shinbashi, Minato-ku, Tokyo 105-0003, Japan
| | - Mai Komori
- VLP Therapeutics, Inc., Gaithersburg, MD 20878, USA
| | | | - Shigeru Suga
- National Hospital Organization, Mie National Hospital, Tsu, Mie 514-0125, Japan
| | | | - Takasuke Fukuhara
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido 060-0815, Japan
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yasumasa Iwatani
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi 460-0001, Japan
- Division of Basic Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Takuya Yamamoto
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Nobuaki Sato
- VLP Therapeutics Japan, Inc., 1-16-4 Nishi-Shinbashi, Minato-ku, Tokyo 105-0003, Japan
| | - Wataru Akahata
- VLP Therapeutics Japan, Inc., 1-16-4 Nishi-Shinbashi, Minato-ku, Tokyo 105-0003, Japan
| |
Collapse
|
39
|
Krasner H, Harmon N, Martin J, Olaco CA, Netski DM, Batra K. Community Level Correlates of COVID-19 Booster Vaccine Hesitancy in the United States: A Cross-Sectional Analysis. Vaccines (Basel) 2024; 12:167. [PMID: 38400150 PMCID: PMC10892894 DOI: 10.3390/vaccines12020167] [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: 12/05/2023] [Revised: 01/27/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Introduction: Evidence exists that individual-level sociodemographic factors contribute to vaccine hesitancy, but it is unknown how community-level factors affect COVID-19 booster dose hesitancy. The current study aims to fill this knowledge gap by comparing data from a nationwide survey on COVID-19 vaccine hesitancy with a community-level indicator, i.e., the Distressed Communities Index (DCI). Methods: Attitudes toward vaccinations, vaccine literacy, COVID-19 vaccine confidence index, and trust were measured using a 48-item, psychometrically valid and reliable survey tool. In this study, 2138 survey participants residing in the United States were divided into quintiles of varying community distress levels based on their zip codes using the DCI. Data were analyzed through Chi-square, one-way ANOVA, and post hoc analysis with Tukey's test. Results: A significantly higher proportion of participants from the distressed communities had lower trust than their prosperous counterparts (26.6% vs. 37.6%, p < 0.001). On the contrary, participants from the prosperous communities had significantly higher vaccine confidence index scores than those in distressed communities (2.22 ± 1.13 vs. 1.70 ± 1.01, p < 0.001). Conclusions: These findings affirm the importance of developing community-level interventions to promote trust in COVID-19 vaccinations and increase booster dose uptake. From these results, future studies can examine the efficacy of various community-level interventions.
Collapse
Affiliation(s)
- Henry Krasner
- Kirk Kerkorian School of Medicine at UNLV, University of Nevada, Las Vegas, NV 89102, USA; (J.M.); (C.-A.O.)
| | - Nicolette Harmon
- Department of Epidemiology and Biostatistics, School of Public Health, University of Nevada, Las Vegas, NV 89119, USA;
| | - Jeffrey Martin
- Kirk Kerkorian School of Medicine at UNLV, University of Nevada, Las Vegas, NV 89102, USA; (J.M.); (C.-A.O.)
| | - Crysty-Ann Olaco
- Kirk Kerkorian School of Medicine at UNLV, University of Nevada, Las Vegas, NV 89102, USA; (J.M.); (C.-A.O.)
| | - Dale M. Netski
- Office of Faculty Affairs, Kirk Kerkorian School of Medicine at UNLV, University of Nevada, Las Vegas, NV 89102, USA;
- Department of Medical Education, Kirk Kerkorian School of Medicine at UNLV, University of Nevada, Las Vegas, NV 89102, USA
| | - Kavita Batra
- Department of Medical Education, Kirk Kerkorian School of Medicine at UNLV, University of Nevada, Las Vegas, NV 89102, USA
- Office of Research, Kirk Kerkorian School of Medicine at UNLV, University of Nevada, Las Vegas, NV 89102, USA
| |
Collapse
|
40
|
Lo E, Brousseau N, Defay F, Fortin É, Kiely M. Neighborhood-level vaccine impact on COVID-19 infection and hospital admission in Quebec, Canada, during the Delta and early Omicron periods. Vaccine 2024; 42:636-644. [PMID: 38135643 DOI: 10.1016/j.vaccine.2023.12.043] [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: 07/11/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
OBJECTIVE To assess the impact of COVID-19 vaccination on COVID-19 infection and hospitalisation at the population-level, and to assess the indirect effects of vaccination in the province of Quebec, Canada. METHODS We performed a time-stratified, neighborhood-level ecologic study. The exposure was neighborhood-level vaccination (primary series) coverage; outcomes were COVID-19 infection and hospitalisation rates. We used robust Poisson regression to estimate weekly relative rates of infection and hospitalisation versus vaccination. RESULTS Higher vaccination coverage was associated with lower COVID-19 infection rates from July 18-December 4 for the year 2021 (Delta period) (RR≈0.46 [0.39; 0.54] - 0.94 [0.83; 1.05], 85-100% vs. 60-74% coverage). From December 5-December 25, this association reversed (RR≈1.28 [1.16; 1.41] - 1.41 [1.31; 1.52]), possibly due to the Omicron variant, social behaviors and accumulation of susceptibles in more vaccinated neighborhoods. Vaccine impact against hospitalisation was maintained throughout (RR≈0.43 [0.29; 0.65] - 0.88 [0.64; 1.22]). Vaccination provided substantial indirect protection (RR≈0.43 [0.34; 0.54] - 0.81 [0.65; 1.03]). CONCLUSIONS This study confirmed the protective impact of vaccination against severe disease regardless of variant, at the population level. Ecological analyses are a valuable strategy to evaluate vaccination programs. Population-level effects can have substantial effects and should be accounted for in public health and vaccination program planning.
Collapse
Affiliation(s)
- Ernest Lo
- Institut national de santé publique du Québec (INSPQ), Québec, QC, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada.
| | - Nicholas Brousseau
- Institut national de santé publique du Québec (INSPQ), Québec, QC, Canada; Département de médecine sociale et préventive, Université Laval, Québec, QC, Canada; Centre de Recherche du CHU de Québec - Université Laval, Québec, QC, Canada.
| | - Fannie Defay
- Institut national de santé publique du Québec (INSPQ), Québec, QC, Canada.
| | - Élise Fortin
- Institut national de santé publique du Québec (INSPQ), Québec, QC, Canada; Département de médecine sociale et préventive, Université Laval, Québec, QC, Canada; Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, Canada.
| | - Marilou Kiely
- Institut national de santé publique du Québec (INSPQ), Québec, QC, Canada; Centre de Recherche du CHU de Québec - Université Laval, Québec, QC, Canada.
| |
Collapse
|
41
|
Di Chiara C, Cantarutti A, Raffaella Petrara M, Bonfante F, Benetti E, Boracchini R, Bosa L, Carmona F, Cosma C, Cotugno N, Le Prevost M, Martini G, Meneghel A, Pagliari M, Palma P, Ruffoni E, Zin A, De Rossi A, Giaquinto C, Donà D, Padoan A. Stronger and durable SARS-CoV-2 immune response to mRNA vaccines in 5-11 years old children with prior COVID-19. Vaccine 2024; 42:263-270. [PMID: 38071105 DOI: 10.1016/j.vaccine.2023.12.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/08/2023] [Accepted: 12/01/2023] [Indexed: 01/01/2024]
Abstract
BACKGROUND AND OBJECTIVES mRNA vaccines elicit a durable humoral response to SARS-CoV-2 in adults, whereas evidence in children is scarce. This study aimed to assess the early and long-term immune response to the mRNA vaccine in children with or without previous SARS-CoV-2 infection. METHODS In a multicentre prospective observational study, we profiled the immune response to the Pfizer BioNTech (BNT162b2) vaccine in 5-11-year-old children attending the University Pediatric Hospital of Padua and Bambino-Gesù Hospital in Rome (Italy) from December-2021 to February-2023. Blood samples were collected pre-, 1-, and 6-months after vaccination. Neutralizing antibodies (NAbs) and anti-spike-receptor-binding-domain (anti-S-RBD) IgG titers were analyzed through Plaque Reduction Neutralization Test (PRNT) and chemiluminescent immune-enzymatic assay (CLIA), respectively. Immune cell phenotypes were analyzed by flow cytometry. RESULTS Sixty children (26 [43 %] female, median age = 8 years [IQR = 7-10.7]) were enrolled in the study, including 46 children with a laboratory-confirmed previous COVID-19 (SARS-CoV-2-recovered) and 14 SARS-CoV-2-naïve participants defined as the absence of antigen-specific antibodies before vaccination. SARS-CoV-2-recovered participants recorded higher anti-S-RBD IgG and Wild-type and Omicron BA.2 NAbs titers than SARS-CoV-2-naïve participants at both 1- and 6-months after vaccination. Antibody titers correlated with T (Tregs) and B (Bregs) regulatory cell frequencies in SARS-CoV-2-recovered children. Both SARS-CoV-2-recovered and SARS-CoV-2-naïve participants decreased antibody titers by approximately 100 to 250 % from 1 to 6 months. While children with immunocompromising underlying conditions developed immune responses comparable to those of healthy children, solid organ transplant recipients exhibited lower levels of NAbs and anti-S-RBD IgG titers, as well as reduced frequencies of Tregs and Bregs. CONCLUSIONS mRNA vaccination triggered a higher production of specific anti-SARS-CoV-2 antibodies along with increased levels of regulatory cells in children with previous SARS-CoV-2 infection up to the following 6 months. These findings provide insights into boosting pre-existing immunity.
Collapse
Affiliation(s)
- Costanza Di Chiara
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy; Penta - Child Health Research, Corso Stati Uniti, 4 - 35127 Padua, Italy.
| | - Anna Cantarutti
- Department of Statistics and Quantitative Methods, Division of Biostatistics, Epidemiology and Public Health, Laboratory of Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, Piazza dell'Ateneo Nuovo, 1 - 20126 Milan, Italy.
| | - Maria Raffaella Petrara
- Oncology and Immunology Section, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani, 2 - 35124 Padua, Italy.
| | - Francesco Bonfante
- Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell'Università, 10 - 35020 Legnaro (Padua), Italy.
| | - Elisa Benetti
- Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padua, Italy.
| | - Riccardo Boracchini
- Department of Statistics and Quantitative Methods, Division of Biostatistics, Epidemiology and Public Health, Laboratory of Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, Piazza dell'Ateneo Nuovo, 1 - 20126 Milan, Italy.
| | - Luca Bosa
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy.
| | - Francesco Carmona
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64 - 35128 Padua, Italy.
| | - Chiara Cosma
- Department of Laboratory Medicine, University-Hospital of Padova, Via Giambattista Belzoni, 160 - 35121 Padua, Italy.
| | - Nicola Cotugno
- Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Systems Medicine, University of Rome "Tor Vergata", Piazza Sant'Onofrio, 4 - 00165 Rome, Italy.
| | - Marthe Le Prevost
- Medical Research Council Clinical Trials Unit at University College London, 90 High Holborn, WC1V 6LJ London, United Kingdom.
| | - Giorgia Martini
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy.
| | - Alessandra Meneghel
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy.
| | - Matteo Pagliari
- Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell'Università, 10 - 35020 Legnaro (Padua), Italy.
| | - Paolo Palma
- Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Systems Medicine, University of Rome "Tor Vergata", Piazza Sant'Onofrio, 4 - 00165 Rome, Italy.
| | - Elena Ruffoni
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64 - 35128 Padua, Italy.
| | - Annachiara Zin
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy.
| | - Anita De Rossi
- Oncology and Immunology Section, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani, 2 - 35124 Padua, Italy; Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64 - 35128 Padua, Italy.
| | - Carlo Giaquinto
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy; Penta - Child Health Research, Corso Stati Uniti, 4 - 35127 Padua, Italy.
| | - Daniele Donà
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy; Penta - Child Health Research, Corso Stati Uniti, 4 - 35127 Padua, Italy.
| | - Andrea Padoan
- Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padua, Italy.
| |
Collapse
|
42
|
Wells AU. COVID-19 Vaccine Efficacy Over Time: Severe Disease in Hospitalized Patients. Radiology 2024; 310:e233340. [PMID: 38259212 DOI: 10.1148/radiol.233340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Affiliation(s)
- Athol U Wells
- From the Interstitial Lung Disease Unit, Royal Brompton and Harefield NHS Foundation Trust, London & National Lung & Lung Institute, Imperial College London, Sydney Street, London SW3 6NP, United Kingdom
| |
Collapse
|
43
|
Altorki TA, Abdulal RH, Suliman BA, Aljeraisi TM, Alsharef A, Abdulaal WH, Alfaleh MA, Algaissi AA, Alhabbab RY, Ozbak H, Eid HM, Almutawif YA, Li X, Al-Rabia MW, Zhang Q, Mahmoud AB, Mahallawi WH, Hashem AM. Robust memory humoral immune response to SARS-CoV-2 in the tonsils of adults and children. Front Immunol 2023; 14:1291534. [PMID: 38149243 PMCID: PMC10750384 DOI: 10.3389/fimmu.2023.1291534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/17/2023] [Indexed: 12/28/2023] Open
Abstract
Background Adaptive humoral immunity against SARS-CoV-2 has mainly been evaluated in peripheral blood. Human secondary lymphoid tissues (such as tonsils) contain large numbers of plasma cells that secrete immunoglobulins at mucosal sites. Yet, the role of mucosal memory immunity induced by vaccines or natural infection against SARS-CoV-2 and its variants is not fully understood. Methods Tonsillar mononuclear cells (TMNCs) from adults (n=10) and children (n=11) were isolated and stimulated using positive SARS-CoV-2 nasal swabs. We used endpoint enzyme-linked immunosorbent assays (ELISAs) for the measurement of anti-S1, -RBD, and -N IgG antibody levels and a pseudovirus microneutralization assay to assess neutralizing antibodies (nAbs) in paired serum and supernatants from stimulated TMNCs. Results Strong systemic humoral response in previously SARS-CoV-2 infected and vaccinated adults and children was observed in accordance with the reported history of the participants. Interestingly, we found a significant increase in anti-RBD IgG (305 and 834 folds) and anti-S1 IgG (475 and 443 folds) in the stimulated TMNCs from adults and children, respectively, compared to unstimulated cells. Consistently, the stimulated TMNCs secreted higher levels of nAbs against the ancestral Wuhan strain and the Omicron BA.1 variant compared to unstimulated cells by several folds. This increase was seen in all participants including children with no known history of infection, suggesting that these participants might have been previously exposed to SARS-CoV-2 and that not all asymptomatic cases necessarily could be detected by serum antibodies. Furthermore, nAb levels against both strains were significantly correlated in adults (r=0.8788; p = 0.0008) and children (r = 0.7521; p = 0.0076), and they strongly correlated with S1 and RBD-specific IgG antibodies. Conclusion Our results provide evidence for persistent mucosal humoral memory in tonsils from previously infected and/or vaccinated adults and children against recent and old variants upon re-exposure. They also highlight the importance of targeting mucosal sites with vaccines to help control infection at the primary sites and prevent potential breakthrough infections.
Collapse
Affiliation(s)
- Tarfa A. Altorki
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rwaa H. Abdulal
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Bandar A. Suliman
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Talal M. Aljeraisi
- Otorhinolaryngology, Head and Neck Surgery Department, Faculty of Medicine, Taibah University, Madinah, Saudi Arabia
| | - Asem Alsharef
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Wesam H. Abdulaal
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed A. Alfaleh
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdullah A. Algaissi
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Rowa Y. Alhabbab
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hani Ozbak
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Hamza Mohammed Eid
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Yahya Ahmad Almutawif
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Xuguang Li
- Centre for Oncology and Regulatory Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada and World Health Organization Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Mohammed W. Al-Rabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Qibo Zhang
- Academic and Research Departments, Section of Immunology, School of Biosciences and Medicine University of Surrey, Surrey, United Kingdom
| | - Ahmed Bakur Mahmoud
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
- Health and Life Research Center, Taibah University, Madinah, Saudi Arabia
| | - Waleed H. Mahallawi
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Anwar M. Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
44
|
Towett G, Snead RS, Grigoryan K, Marczika J. Geographical and practical challenges in the implementation of digital health passports for cross-border COVID-19 pandemic management: a narrative review and framework for solutions. Global Health 2023; 19:98. [PMID: 38066568 PMCID: PMC10709942 DOI: 10.1186/s12992-023-00998-7] [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: 08/20/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
The rapid global spread of infectious diseases, epitomized by the recent COVID-19 pandemic, has highlighted the critical need for effective cross-border pandemic management strategies. Digital health passports (DHPs), which securely store and facilitate the sharing of critical health information, including vaccination records and test results, have emerged as a promising solution to enable safe travel and access to essential services and economic activities during pandemics. However, the implementation of DHPs faces several significant challenges, both related to geographical disparities and practical considerations, necessitating a comprehensive approach for successful global adoption. In this narrative review article, we identify and elaborate on the critical geographical and practical barriers that hinder global adoption and the effective utilization of DHPs. Geographical barriers are complex, encompassing disparities in vaccine access, regulatory inconsistencies, differences across countries in data security and users' privacy policies, challenges related to interoperability and standardization, and inadequacies in technological infrastructure and limited access to digital technologies. Practical challenges include the possibility of vaccine contraindications and breakthrough infections, uncertainties surrounding natural immunity, and limitations of standard tests in assessing infection risk. To address geographical disparities and enhance the functionality and interoperability of DHPs, we propose a framework that emphasizes international collaboration to achieve equitable access to vaccines and testing resources. Furthermore, we recommend international cooperation to establish unified vaccine regulatory frameworks, adopting globally accepted standards for data privacy and protection, implementing interoperability protocols, and taking steps to bridge the digital divide. Addressing practical challenges requires a meticulous approach to assessing individual risk and augmenting DHP implementation with rigorous health screenings and personal infection prevention measures. Collectively, these initiatives contribute to the development of robust and inclusive cross-border pandemic management strategies, ultimately promoting a safer and more interconnected global community in the face of current and future pandemics.
Collapse
|
45
|
Mahdi PDBM, Almukhtar DM. Role Of Vaccines Against COVID-19 Pandemic. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:355-357. [PMID: 37473842 DOI: 10.1016/j.slasd.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023]
Abstract
Coronaviruses (CoV) are one of the largest families of viruses that infect human beings causing mild common cold or severe diseases like Middle East Respiratory Syndrome (MERS-CoV), and Severe Acute Respiratory Syndrome (SARS-CoV). A new strain emerged known as novel coronavirus (nCoV) causing fatal respiratory failure disease. This virus was characterized by rapid spread from asymptomatic and symptomatic patients to healthy people. Thus, vaccine should be considered as one of the important protective measures to control the spread of this virus. One of the challenges to this vaccine is the high mutation rate of this virus and appearance of new strains. Therefore, vaccine should stimulate the immune system in order to overcome the emergence of new strain of this virus.
Collapse
Affiliation(s)
- Professor Dr Batool Mutar Mahdi
- Consultant Clinical Immunology, Head of HLA Research Unit, Department of Microbiology, Al-Kindy College of Medicine, University of Baghdad, Baghdad, Iraq.
| | | |
Collapse
|
46
|
Alpizar SA, Accini J, Anderson DC, Eysa B, Medina-Piñón I, Ohmagari N, Ostrovskyy MM, Aggrey-Amable A, Beck K, Byrne D, Grayson S, Hwang PMT, Lonchar JD, Strizki J, Xu Y, Paschke A, De Anda CS, Sears PS. Molnupiravir for intra-household prevention of COVID-19: The MOVe-AHEAD randomized, placebo-controlled trial. J Infect 2023; 87:392-402. [PMID: 37690669 DOI: 10.1016/j.jinf.2023.08.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/23/2023] [Accepted: 08/31/2023] [Indexed: 09/12/2023]
Abstract
OBJECTIVES To evaluate the efficacy and safety of molnupiravir for intra-household post-exposure prophylaxis (PEP) of COVID-19. METHODS MOVe-AHEAD was a randomized, controlled, double-blind, phase 3 trial comparing molnupiravir (800 mg twice daily for 5 days) with placebo. Eligible participants were adult, unvaccinated, asymptomatic household contacts of patients with laboratory-confirmed COVID-19. The primary efficacy endpoint was the incidence of COVID-19 through day 14 in modified intention-to-treat (MITT) participants (those who received ≥1 dose of study intervention) without detectable SARS-CoV-2 at baseline, termed the MITT-VN population. Superiority of molnupiravir was prespecified as a stratified one-sided p-value of <0.0249 for the treatment difference in this endpoint. RESULTS The MITT population comprised 763 participants randomized to molnupiravir and 764 to placebo; 83.6% had anti-SARS-CoV-2 antibodies at baseline. In the MITT-VN population, COVID-19 rates through day 14 were 6.5% with molnupiravir and 8.5% with placebo (one-sided p-value: 0.0848). In the molnupiravir arm, 25/35 of confirmed COVID-19 events (71.4%) occurred after completion of treatment (versus 17/49 [34.7%] for placebo). Adverse event rates were low and similar between molnupiravir and placebo. CONCLUSIONS Molnupiravir was well-tolerated but did not meet the prespecified superiority criterion, possibly influenced in part by the high pre-existing immunity in the trial population.
Collapse
Affiliation(s)
- Sady A Alpizar
- Clinical Research Trials of Florida, 2713 W. Virginia Ave., Tampa 33607, FL, USA.
| | - Jose Accini
- IPS Centro Científico Asistencial, Cra. 45 #85-49, Barranquilla 080020, Colombia.
| | - Duane C Anderson
- Excel Clinical Research LLC, 3059 S Maryland Pkwy., Las Vegas, NV 89109, USA.
| | - Basem Eysa
- National Hepatology and Tropical Medicine Research Institute, 10 Kasr El, Eini St., Fom Al Khalig Sq., Cairo 11796, Egypt.
| | - Isaí Medina-Piñón
- ICARO Investigaciones en Medicina, Calle Ignacio Allende No. 1015, Chihuahua 31000, Mexico.
| | - Norio Ohmagari
- National Center for Global Health and Medicine Hospital, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan.
| | - Mykola M Ostrovskyy
- Regional Phthisiopulmonological Center, 17 Franka St., Ivano-Frankivsk 76018, Ukraine.
| | | | - Karen Beck
- Merck & Co, Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA
| | - Dana Byrne
- Merck & Co, Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA
| | - Staci Grayson
- Merck & Co, Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA
| | - Peggy M T Hwang
- Merck & Co, Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA
| | - Julia D Lonchar
- Merck & Co, Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA
| | - Julie Strizki
- Merck & Co, Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA
| | - Yayun Xu
- Merck & Co, Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA
| | - Amanda Paschke
- Merck & Co, Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA
| | | | - Pamela S Sears
- Merck & Co, Inc., 126 E. Lincoln Ave., Rahway, NJ 07065, USA.
| |
Collapse
|
47
|
Hogan AB, Wu SL, Toor J, Olivera Mesa D, Doohan P, Watson OJ, Winskill P, Charles G, Barnsley G, Riley EM, Khoury DS, Ferguson NM, Ghani AC. Long-term vaccination strategies to mitigate the impact of SARS-CoV-2 transmission: A modelling study. PLoS Med 2023; 20:e1004195. [PMID: 38016000 PMCID: PMC10715640 DOI: 10.1371/journal.pmed.1004195] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 12/12/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Vaccines have reduced severe disease and death from Coronavirus Disease 2019 (COVID-19). However, with evidence of waning efficacy coupled with continued evolution of the virus, health programmes need to evaluate the requirement for regular booster doses, considering their impact and cost-effectiveness in the face of ongoing transmission and substantial infection-induced immunity. METHODS AND FINDINGS We developed a combined immunological-transmission model parameterised with data on transmissibility, severity, and vaccine effectiveness. We simulated Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) transmission and vaccine rollout in characteristic global settings with different population age-structures, contact patterns, health system capacities, prior transmission, and vaccine uptake. We quantified the impact of future vaccine booster dose strategies with both ancestral and variant-adapted vaccine products, while considering the potential future emergence of new variants with modified transmission, immune escape, and severity properties. We found that regular boosting of the oldest age group (75+) is an efficient strategy, although large numbers of hospitalisations and deaths could be averted by extending vaccination to younger age groups. In countries with low vaccine coverage and high infection-derived immunity, boosting older at-risk groups was more effective than continuing primary vaccination into younger ages in our model. Our study is limited by uncertainty in key parameters, including the long-term durability of vaccine and infection-induced immunity as well as uncertainty in the future evolution of the virus. CONCLUSIONS Our modelling suggests that regular boosting of the high-risk population remains an important tool to reduce morbidity and mortality from current and future SARS-CoV-2 variants. Our results suggest that focusing vaccination in the highest-risk cohorts will be the most efficient (and hence cost-effective) strategy to reduce morbidity and mortality.
Collapse
Affiliation(s)
- Alexandra B. Hogan
- School of Population Health, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Sean L. Wu
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, United States of America
| | - Jaspreet Toor
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Daniela Olivera Mesa
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Patrick Doohan
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Oliver J. Watson
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Peter Winskill
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Giovanni Charles
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Gregory Barnsley
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Eleanor M. Riley
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - David S. Khoury
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Neil M. Ferguson
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Azra C. Ghani
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| |
Collapse
|
48
|
Kieber-Emmons T. Where Are We with COVID Boosters? Monoclon Antib Immunodiagn Immunother 2023; 42:151-152. [PMID: 37862051 DOI: 10.1089/mab.2023.29014.editorial] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
|
49
|
Canuti M, Monti MC, Bobbio C, Muscatello A, Muheberimana T, Baldi SL, Blasi F, Canetta C, Costantino G, Nobili A, Peyvandi F, Tettamanti M, Villa S, Aliberti S, Raviglione MC, Gori A, Bandera A. The role of immune suppression in COVID-19 hospitalization: clinical and epidemiological trends over three years of SARS-CoV-2 epidemic. Front Med (Lausanne) 2023; 10:1260950. [PMID: 37746083 PMCID: PMC10513414 DOI: 10.3389/fmed.2023.1260950] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
Specific immune suppression types have been associated with a greater risk of severe COVID-19 disease and death. We analyzed data from patients >17 years that were hospitalized for COVID-19 at the "Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico" in Milan (Lombardy, Northern Italy). The study included 1727 SARS-CoV-2-positive patients (1,131 males, median age of 65 years) hospitalized between February 2020 and November 2022. Of these, 321 (18.6%, CI: 16.8-20.4%) had at least one condition defining immune suppression. Immune suppressed subjects were more likely to have other co-morbidities (80.4% vs. 69.8%, p < 0.001) and be vaccinated (37% vs. 12.7%, p < 0.001). We evaluated the contribution of immune suppression to hospitalization during the various stages of the epidemic and investigated whether immune suppression contributed to severe outcomes and death, also considering the vaccination status of the patients. The proportion of immune suppressed patients among all hospitalizations (initially stable at <20%) started to increase around December 2021, and remained high (30-50%). This change coincided with an increase in the proportions of older patients and patients with co-morbidities and with a decrease in the proportion of patients with severe outcomes. Vaccinated patients showed a lower proportion of severe outcomes; among non-vaccinated patients, severe outcomes were more common in immune suppressed individuals. Immune suppression was a significant predictor of severe outcomes, after adjusting for age, sex, co-morbidities, period of hospitalization, and vaccination status (OR: 1.64; 95% CI: 1.23-2.19), while vaccination was a protective factor (OR: 0.31; 95% IC: 0.20-0.47). However, after November 2021, differences in disease outcomes between vaccinated and non-vaccinated groups (for both immune suppressed and immune competent subjects) disappeared. Since December 2021, the spread of the less virulent Omicron variant and an overall higher level of induced and/or natural immunity likely contributed to the observed shift in hospitalized patient characteristics. Nonetheless, vaccination against SARS-CoV-2, likely in combination with naturally acquired immunity, effectively reduced severe outcomes in both immune competent (73.9% vs. 48.2%, p < 0.001) and immune suppressed (66.4% vs. 35.2%, p < 0.001) patients, confirming previous observations about the value of the vaccine in preventing serious disease.
Collapse
Affiliation(s)
- Marta Canuti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), Università degli Studi di Milano, Milan, Italy
- Coordinate Research Centre EpiSoMI (Epidemiology and Molecular Surveillance of Infections), Università degli Studi di Milano, Milan, Italy
| | - Maria Cristina Monti
- Department of Public Health, Experimental and Forensic Medicine, Unit of Biostatistics and Clinical Epidemiology, Università degli Studi di Pavia, Pavia, Italy
| | - Chiara Bobbio
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Antonio Muscatello
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | | | - Sante Leandro Baldi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), Università degli Studi di Milano, Milan, Italy
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Ciro Canetta
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Giorgio Costantino
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Alessandro Nobili
- Department of Health Policy, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Flora Peyvandi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Mauro Tettamanti
- Department of Health Policy, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Simone Villa
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), Università degli Studi di Milano, Milan, Italy
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Respiratory Unit, Milan, Italy
| | - Mario C. Raviglione
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), Università degli Studi di Milano, Milan, Italy
| | - Andrea Gori
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), Università degli Studi di Milano, Milan, Italy
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Alessandra Bandera
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| |
Collapse
|
50
|
Chuang YM, Alameh MG, Abouneameh S, Raduwan H, Ledizet M, Weissman D, Fikrig E. A mosquito AgTRIO mRNA vaccine contributes to immunity against malaria. NPJ Vaccines 2023; 8:88. [PMID: 37286568 PMCID: PMC10244833 DOI: 10.1038/s41541-023-00679-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/16/2023] [Indexed: 06/09/2023] Open
Abstract
Malaria begins when an infected mosquito injects saliva containing Plasmodium sporozoites into the skin of a vertebrate host. To prevent malaria, vaccination is the most effective strategy and there is an urgent need for new strategies to enhance current pathogen-based vaccines. Active or passive immunization against a mosquito saliva protein, AgTRIO, contributes to protection against Plasmodium infection of mice. In this study, we generated an AgTRIO mRNA-lipid nanoparticle (LNP) and assessed its potential usefulness as a vaccine against malaria. Immunization of mice with an AgTRIO mRNA-LNP generated a robust humoral response, including AgTRIO IgG2a isotype antibodies that have been associated with protection. AgTRIO mRNA-LNP immunized mice exposed to Plasmodium berghei-infected mosquitoes had markedly reduced initial Plasmodium hepatic infection levels and increased survival compared to control mice. In addition, as the humoral response to AgTRIO waned over 6 months, additional mosquito bites boosted the AgTRIO IgG titers, including IgG1 and IgG2a isotypes, which offers a unique advantage compared to pathogen-based vaccines. These data will aid in the generation of future malaria vaccines that may include both pathogen and vector antigens.
Collapse
Affiliation(s)
- Yu-Min Chuang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Mohamad-Gabriel Alameh
- Institute for RNA Innovation and Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Selma Abouneameh
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Hamidah Raduwan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | - Drew Weissman
- Institute for RNA Innovation and Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|