1
|
Dari A, Pérez Ruixo JJ, Le Gars M, Struyf F, Jacqmin P. Modelling antibody dynamics in humans after different Ad26.COV2.S vaccination schemes. Br J Clin Pharmacol 2025; 91:397-408. [PMID: 39327825 DOI: 10.1111/bcp.16251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/12/2024] [Accepted: 07/27/2024] [Indexed: 09/28/2024] Open
Abstract
AIMS To develop a semimechanistic model that describes the kinetic profile and variability of antibody (Ab) concentrations following vaccination with Ad26.COV2.S at different doses and dosing intervals. METHODS Data were collected from participants randomized into 5 clinical trials receiving the Ad26.COV2.S vaccine. The model considered key elements of humoral immune response, dose proportionality and the evolutionary processes of the immune response. Interindividual variability and covariates were explored. RESULTS Fast and slow kinetic phases of Ab and their evolution over time were differentiated. After first and second administrations, Ab concentrations of both phases increased less than dose proportionally, indicating a saturation of B-cell production processes. Ab concentrations produced during the fast kinetic phase increased significantly after the second administration, indicating an underlying evolutive process after antigen exposures. For the slow kinetic phase, a less pronounced increase occurred after the second and third administrations but was relatively higher in subjects who had low concentrations after the first administration. Ab concentrations of the slow phase were higher in females and decreased with age. After multiple administrations, the fast phase had Ab maximum concentrations about 5 times higher than the slow phase. The limiting kinetic factors in the fast and slow phases were the elimination rates of Ab itself and Ab producing cells, respectively. CONCLUSION The model appears suitable to quantitatively describe the inter- and intraindividual kinetics of the immune response and the impact of covariates after multiple administrations of a vaccine.
Collapse
Affiliation(s)
- Anna Dari
- Janssen Research & Development, Beerse, Belgium
| | | | | | | | | |
Collapse
|
2
|
Jarras H, Blais I, Goyer B, Bazié WW, Rabezanahary H, Thériault M, Santerre K, Langlois MA, Masson JF, Pelletier JN, Brousseau N, Boudreau D, Trottier S, Baz M, Gilbert C. Impact of SARS-CoV-2 vaccination and of seasonal variations on the innate immune inflammatory response. Front Immunol 2025; 15:1513717. [PMID: 39877354 PMCID: PMC11772892 DOI: 10.3389/fimmu.2024.1513717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction The innate immune response is an important first checkpoint in the evolution of an infection. Although adaptive immunity is generally considered the immune component that retains antigenic memory, innate immune responses can also be affected by previous stimulations. This study evaluated the impact of vaccination on innate cell activation by TLR7/8 agonist R848, as well as seasonal variations. Methods To this end, blood samples from a cohort of 304 food and retail workers from the Quebec City region were collected during three visits at 12-week intervals. Peripheral blood mononuclear cells and polymorphonuclear neutrophils were isolated during the first and third visits and were stimulated with R848 to assess the innate immune response. Results Our results show that IL-8 production after stimulation decreased after vaccination. In addition, the IL-8 response was significantly different depending on the season when the visit occurred, for both COVID-19 vaccinated and unvaccinated individuals. Discussion This study highlights that innate immune responses can be affected by SARS-CoV-2 vaccination and fluctuate seasonally.
Collapse
Affiliation(s)
- Hend Jarras
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Isalie Blais
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Benjamin Goyer
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Wilfried W. Bazié
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Programme de Recherche sur les Maladies Infectieuses, Centre Muraz, Institut National de Santé Publique, Bobo-Dioulasso, Houet, Burkina Faso
| | - Henintsoa Rabezanahary
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Mathieu Thériault
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Kim Santerre
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jean-François Masson
- Department of Chemistry, Institut Courtois, Quebec Center for Advanced Materials, Regroupement québécois sur les matériaux de pointe, and Centre Interdisciplinaire de Recherche sur le Cerveau et l’Apprentissage, Université de Montréal, Montreal, QC, Canada
| | - Joelle N. Pelletier
- Department of Chemistry, Department of Biochemistry, Université de Montréal, Montreal, QC, Canada
- PROTEO — The Québec Network for Research on Protein Function, Engineering, and Applications, Quebec City, QC, Canada
| | - Nicholas Brousseau
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Biological Risks Department, Institut national de santé publique du Québec, Quebec City, QC, Canada
| | - Denis Boudreau
- Département de Chimie et Center for Optics, Photonics and Lasers (COPL), Université Laval, Quebec City, QC, Canada
| | - Sylvie Trottier
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Département de Microbiologie-Infectiologie et d’Immunologie, Faculté de Médecine, Université Laval, Quebec City, QC, Canada
| | - Mariana Baz
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Département de Microbiologie-Infectiologie et d’Immunologie, Faculté de Médecine, Université Laval, Quebec City, QC, Canada
| | - Caroline Gilbert
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Département de Microbiologie-Infectiologie et d’Immunologie, Faculté de Médecine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
3
|
Ibrahimová M, Jamriková V, Pavelková K, Bořecká K. Nucleocapsid Antibodies as an Optimal Serological Marker of SARS-CoV-2 Infection: A Longitudinal Study at the Thomayer University Hospital. J Clin Lab Anal 2025:e25149. [PMID: 39760288 DOI: 10.1002/jcla.25149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 12/04/2024] [Accepted: 12/24/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND The longitudinal study was conducted over the initial 2 years of the COVID-19 pandemic, spanning from June 2020 to December 2022, in healthcare workers (HCWs) of the Thomayer University Hospital. A total of 3892 blood samples were collected and analyzed for total nucleocapsid (N) antibodies. The aim of the study was to evaluate the dynamics of N antibodies, their relationship to the PCR test, spike (S) antibodies, interferon-gamma, and prediction of reinfection with SARS-CoV-2. METHODS Blood collections were performed in three rounds, along with questionnaires addressing clinical symptoms of past infection, PCR testing, and vaccination. Antibody measurements included total N antibodies (Roche Diagnostics) and postvaccination S antibodies (Euroimmun). Cellular immunity was tested by interferon-gamma release assay (Euroimmun). RESULTS At the end of the study, 35.9% of HCWs were positive for N antibodies, and 39.5% of HCWs had either known PCR positivity or N antibodies or both. Ten percent of participants had no knowledge of a COVID-19 infection and 35% of positive individuals exhibited no symptoms. The values of positive antibodies decrease over a period of 6 months to 1 year, depending on the initial value, and their dynamics are highly variable. The study also demonstrated that the highest levels of spike antibodies and interferon-gamma occur during so-called hybrid immunity. CONCLUSION Nucleocapsid antibodies proved valuable in monitoring SARS-CoV-2 infection dynamics, and they may detect cases of SARS-CoV-2 infection missed by PCR tests. The study identified distinct patterns in antibody dynamics and protection of hybrid immunity during reinfection.
Collapse
Affiliation(s)
- Markéta Ibrahimová
- Laboratory of Immunology, Thomayer University Hospital, Prague, Czech Republic
| | - Vladislava Jamriková
- Department of Clinical Biochemistry, Thomayer University Hospital, Prague, Czech Republic
| | - Kateřina Pavelková
- Department of Hospital Epidemiology and Infection Control, Thomayer University Hospital, Prague, Czech Republic
| | - Klára Bořecká
- Department of Clinical Biochemistry, Thomayer University Hospital, Prague, Czech Republic
| |
Collapse
|
4
|
Blanco J, Trinité B, Puig‐Barberà J. Rethinking Optimal Immunogens to Face SARS-CoV-2 Evolution Through Vaccination. Influenza Other Respir Viruses 2025; 19:e70076. [PMID: 39871737 PMCID: PMC11773156 DOI: 10.1111/irv.70076] [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: 08/26/2024] [Revised: 12/23/2024] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
SARS-CoV-2, which originated in China in late 2019, quickly fueled the global COVID-19 pandemic, profoundly impacting health and the economy worldwide. A series of vaccines, mostly based on the full SARS-CoV-2 Spike protein, were rapidly developed, showing excellent humoral and cellular responses and high efficacy against both symptomatic infection and severe disease. However, viral evolution and the waning humoral neutralizing responses strongly challenged vaccine long term effectiveness, mainly against symptomatic infection, making necessary a strategy of repeated and updated booster shots. In this repeated vaccination context, antibody repertoire diversification was evidenced, although immune imprinting after booster doses or reinfection was also demonstrated and identified as a major determinant of immunological responses to repeated antigen exposures. Considering that a small domain of the SARS-CoV-2 Spike protein, the receptor binding domain (RBD), is the major target of neutralizing antibodies and concentrates most viral mutations, the following text aims to provide insights into the ongoing debate over the best strategies for vaccine boosters. We address the relevance of developing new booster vaccines that target the evolving RBD, thus focusing on the relevant antigenic sites of the SARS-CoV-2 new variants. A combination of this strategy with immunofusing and computerized approaches could minimize immune imprinting, therefore optimizing neutralizing immune responses and booster vaccine efficacy.
Collapse
Affiliation(s)
- Julià Blanco
- IrsiCaixaBadalonaCataloniaSpain
- Germans Trias i Pujol Research Institute (IGTP)BadalonaCataloniaSpain
- CIBER de Enfermedades InfecciosasMadridSpain
- Chair in Infectious Diseases and Immunity, Faculty of MedicineUniversity of Vic‐Central University of Catalonia (UVic‐UCC)VicCataloniaSpain
| | | | - Joan Puig‐Barberà
- Área de Investigación en VacunasFundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat ValencianaValenciaSpain
| |
Collapse
|
5
|
Nersesjan V, Boldingh MI, Paulsen EQ, Argren M, Høgestøl E, Aamodt AH, Popperud TH, Kondziella D, Jørgensen CS, Jensen VVS, Benros ME. Antibodies against SARS-CoV-2 spike protein in the cerebrospinal fluid of COVID-19 patients and vaccinated controls: a multicentre study. J Neurol 2024; 272:60. [PMID: 39680178 DOI: 10.1007/s00415-024-12769-4] [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: 09/09/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 12/17/2024]
Abstract
INTRODUCTION SARS-CoV-2 antibodies in the cerebrospinal fluid (CSF) of COVID-19 patients possibly reflect blood-cerebrospinal fluid barrier (BCB) disruption due to systemic inflammation. However, some studies indicate that CSF antibodies signal a neurotropic infection. Currently, larger studies are needed to clarify this, and it is unknown if CSF antibodies appear solely after infection or also after COVID-19 vaccination. Therefore, we aimed to investigate the CSF dynamics of SARS-CoV-2 antibodies in a multicenter study of COVID-19 patients and vaccinated controls. METHODS A cohort study of Danish and Norwegian COVID-19 patients and controls investigated with a lumbar puncture (April 2020-December 2022). Serum and CSF were analysed locally for routine investigations, and centrally at Statens Serum Institut (Danish governmental public health institute) for SARS-CoV-2 IgG antibodies against the spike protein using the Euroimmun (quantitative) and Wantai (qualitative) assays. Primary outcome was the quantity of CSF SARS-CoV-2 antibodies post-COVID versus post-vaccination. Secondary outcomes included regression models examining the relationship between CSF antibodies and serum levels, albumin ratio, CSF pleocytosis, COVID-19 severity, and temporal antibody dynamics. RESULTS We included 124 individuals (Mean [SD] age 47.2 [16.6]; 59.7% males surviving COVID-19 and controls. Of these, 86 had paired CSF-serum testing. Antibody-index calculations did not support a SARS-CoV-2 brain infection. Multi-variate regression revealed that CSF SARS-CoV-2 antibodies were most strongly influenced by serum antibody levels and BCB permeability, as measured by increasing albumin ratio. CSF antibody levels displayed a dose-response relationship (p < 0.0001) influenced by preceding vaccinations or infections. CSF antibody levels (median [IQR]) were highest among those both previously infected and vaccinated, 100.0 [25.0-174.0], and those vaccinated without prior infection, 85.0 [12.0-142.0], and lowest among previously infected individuals without preceding vaccination, 5.9 [2.7-55.1], (p = 0.003). SARS-CoV-2 antibodies in CSF were also detected via qualitative assays in the COVID-19 (46.8%) and vaccinated (78.6%) groups, p = 0.03. CONCLUSION SARS-CoV-2 antibodies detected in CSF can be derived following both infection and vaccination for COVID-19. CSF antibody levels increase in a dose-response relationship with the number of prior infections and vaccinations and are most strongly influenced by serum antibody levels and BCB permeability. These findings stress the importance of carefully interpreting CSF antibody results when assessing neurological complications following infections not categorized as neurotropic.
Collapse
Affiliation(s)
- Vardan Nersesjan
- Copenhagen Research Center for Biological and Precision Psychiatry, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | | | - Maria Argren
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Einar Høgestøl
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Department of Psychology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne Hege Aamodt
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Department of Neuromedicine and Movement Science, The Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Daniel Kondziella
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Charlotte Sværke Jørgensen
- Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| | | | - Michael E Benros
- Copenhagen Research Center for Biological and Precision Psychiatry, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Bayarri-Olmos R, Sutta A, Rosbjerg A, Mortensen MM, Helgstrand C, Nielsen PF, Pérez-Alós L, González-García B, Johnsen LB, Matthiesen F, Egebjerg T, Hansen CB, Sette A, Grifoni A, Antunes RDS, Garred P. Unraveling the impact of SARS-CoV-2 mutations on immunity: insights from innate immune recognition to antibody and T cell responses. Front Immunol 2024; 15:1412873. [PMID: 39720734 PMCID: PMC11666439 DOI: 10.3389/fimmu.2024.1412873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 11/22/2024] [Indexed: 12/26/2024] Open
Abstract
Throughout the COVID-19 pandemic, the emergence of new viral variants has challenged public health efforts, often evading antibody responses generated by infections and vaccinations. This immune escape has led to waves of breakthrough infections, raising questions about the efficacy and durability of immune protection. Here we focus on the impact of SARS-CoV-2 Delta and Omicron spike mutations on ACE-2 receptor binding, protein stability, and immune response evasion. Delta and Omicron variants had 3-5 times higher binding affinities to ACE-2 than the ancestral strain (KDwt = 23.4 nM, KDDelta = 8.08 nM, KDBA.1 = 4.77 nM, KDBA.2 = 4.47 nM). The pattern recognition molecule mannose-binding lectin (MBL) has been shown to recognize the spike protein. Here we found that MBL binding remained largely unchanged across the variants, even after introducing mutations at single glycan sites. Although MBL binding decreased post-vaccination, it increased by 2.6-fold upon IgG depletion, suggesting a compensatory or redundant role in immune recognition. Notably, we identified two glycan sites (N717 and N801) as potentially essential for the structural integrity of the spike protein. We also evaluated the antibody and T cell responses. Neutralization by serum immunoglobulins was predominantly mediated by IgG rather than IgA and was markedly impaired against the Delta (5.8-fold decrease) and Omicron variants BA.1 (17.4-fold) and BA.2 (14.2-fold). T cell responses, initially conserved, waned rapidly within 3 months post-Omicron infection. Our data suggests that immune imprinting may have hindered antibody and T cell responses toward the variants. Overall, despite decreased antibody neutralization, MBL recognition and T cell responses were generally unaffected by the variants. These findings extend our understanding of the complex interplay between viral adaptation and immune response, underscoring the importance of considering MBL interactions, immune imprinting, and viral evolution dynamics in developing new vaccine and treatment strategies.
Collapse
Affiliation(s)
- Rafael Bayarri-Olmos
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Recombinant Protein and Antibody Unit, Copenhagen University Hospital,
Rigshospitalet, Copenhagen, Denmark
| | - Adrian Sutta
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Recombinant Protein and Antibody Unit, Copenhagen University Hospital,
Rigshospitalet, Copenhagen, Denmark
| | - Anne Rosbjerg
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Recombinant Protein and Antibody Unit, Copenhagen University Hospital,
Rigshospitalet, Copenhagen, Denmark
| | | | | | | | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Beatriz González-García
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | - Cecilie Bo Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, United States
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | | | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Odle A, Kar M, Verma AK, Sariol A, Meyerholz DK, Suthar MS, Wong LYR, Perlman S. Tissue-resident memory T cells contribute to protection against heterologous SARS-CoV-2 challenge. JCI Insight 2024; 9:e184074. [PMID: 39405115 PMCID: PMC11623939 DOI: 10.1172/jci.insight.184074] [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: 06/18/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
New vaccine formulations are based on circulating strains of virus, which have tended to evolve to more readily transmit human to human and to evade the neutralizing antibody response. An assumption of this approach is that ancestral strains of virus will not recur. Recurrence of these strains could be a problem for individuals not previously exposed to ancestral spike protein. Here, we addressed this by infecting mice with recent SARS-CoV-2 variants and then challenging them with a highly pathogenic mouse-adapted virus closely related to the ancestral Wuhan-1 strain (SARS2-N501YMA30). We found that challenged mice were protected from severe disease, despite having low or no neutralizing antibodies against SARS2-N501YMA30. T cell depletion from previously infected mice did not diminish infection against clinical disease, although it resulted in delayed virus clearance in the nasal turbinate and, in some cases, in the lungs. Levels of tissue-resident memory T cells were significantly elevated in the nasal turbinate of previously infected mice compared with that of naive mice. However, this phenotype was not seen in lung tissues. Together, these results indicate that the immune response to newly circulating variants afforded protection against reinfection with the ancestral virus that was in part T cell based.
Collapse
Affiliation(s)
- Abby Odle
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Meenakshi Kar
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Vaccine Center, Atlanta, Georgia, USA
| | - Abhishek K. Verma
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Alan Sariol
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Mehul S. Suthar
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Vaccine Center, Atlanta, Georgia, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lok-Yin Roy Wong
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
- Department of Microbiology, Biochemistry and Molecular Genetics and
- Center for Virus-Host-Innate Immunity, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
8
|
Szachniewicz MM, Neustrup MA, van den Eeden SJF, van Meijgaarden KE, Franken KLMC, van Veen S, Koning RI, Limpens RWAL, Geluk A, Bouwstra JA, Ottenhoff THM. Evaluation of PLGA, lipid-PLGA hybrid nanoparticles, and cationic pH-sensitive liposomes as tuberculosis vaccine delivery systems in a Mycobacterium tuberculosis challenge mouse model - A comparison. Int J Pharm 2024; 666:124842. [PMID: 39424087 DOI: 10.1016/j.ijpharm.2024.124842] [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: 03/11/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Tuberculosis (TB) continues to pose a global threat for millennia, currently affecting over 2 billion people and causing 10.6 million new cases and 1.3 million deaths annually. The only existing vaccine, Mycobacterium Bovis Bacillus Calmette-Guérin (BCG), provides highly variable and inadequate protection in adults and adolescents. This study explores newly developed subunit tuberculosis vaccines that use a multistage protein fusion antigen Ag85b-ESAT6-Rv2034 (AER). The protection efficacy, as well as in vivo induced immune responses, were compared for five vaccines: BCG; AER-CpG/MPLA mix; poly(D,L-lactic-co-glycolic acid) (PLGA); lipid-PLGA hybrid nanoparticles (NPs); and cationic pH-sensitive liposomes (the latter three delivering AER together with CpG and MPLA). All vaccines, except the AER-adjuvant mix, induced protection in Mycobacterium tuberculosis (Mtb)-challenged C57/Bl6 mice as indicated by a significant reduction in bacterial burden in lungs and spleens of the animals. Four AER-based vaccines significantly increased the number of circulating multifunctional CD4+ and CD8+ T-cells producing IL-2, IFNγ, and TNFα, exhibiting a central memory phenotype. Furthermore, AER-based vaccines induced an increase in CD69+ B-cell counts as well as high antigen-specific antibody titers. Unexpectedly, none of the observed immune responses were associated with the bacterial burden outcome, such that the mechanism responsible for the observed vaccine-induced protection of these vaccines remains unclear. These findings suggest the existence of non-classical protective mechanisms for Mtb infection, which could, once identified, provide interesting targets for novel vaccines.
Collapse
Affiliation(s)
- Mikołaj M Szachniewicz
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands.
| | - Malene A Neustrup
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, the Netherlands
| | - Susan J F van den Eeden
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Krista E van Meijgaarden
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Kees L M C Franken
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Suzanne van Veen
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Roman I Koning
- Electron Microscopy Facility, Leiden University Medical Center (LUMC), the Netherlands
| | - Ronald W A L Limpens
- Electron Microscopy Facility, Leiden University Medical Center (LUMC), the Netherlands
| | - Annemieke Geluk
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Joke A Bouwstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, the Netherlands
| | - Tom H M Ottenhoff
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| |
Collapse
|
9
|
Harrache A, Saker K, Mokdad B, Generenaz L, Saade C, Pons S, Fassier JB, Bal A, Trabaud MA, Rabilloud M, Abichou-Klich A, Trouillet-Assant S. Anti-RBD IgG dynamics following infection or vaccination. Vaccine 2024; 42:126464. [PMID: 39432992 DOI: 10.1016/j.vaccine.2024.126464] [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: 04/12/2024] [Revised: 09/10/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
Identifying parameters influencing SARS-CoV-2 antibody dynamics post infection or vaccination is crucial for refining vaccination strategies. In a longitudinal analysis of 1340 samples from 375 healthcare workers, we characterized peak serological response and IgG half-life. Peak antibody titers post 2 vaccine doses were ∼ 20 times higher than natural infection; conversely, infected individuals had extended antibody half-life. Clinical and demographical factors such as BMI, age and smoking shaped peak response without affecting anti-RBD IgG half-life. A third mRNA vaccine dose increased peak antibody titers and prolonged half-life compared to the second dose. These findings underscore the diverse kinetics of SARS-CoV-2 antibody responses, which is influenced by immunization type/number and clinical factors.
Collapse
Affiliation(s)
- Amira Harrache
- Biostatistics Department, Hospices Civils de Lyon, Lyon France. CNRS, UMR 5558, University of Lyon, Biometrics and Evolutionary Biology Laboratory, Biostatistics-Health Team, 69100 Villeurbanne, France
| | - Kahina Saker
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Bouchra Mokdad
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Laurence Generenaz
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Carla Saade
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Sylvie Pons
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Jean-Baptiste Fassier
- Occupational Health and Medicine Department, Hospices Civils de Lyon, 69002 Lyon, France
| | - Antonin Bal
- Virology Laboratory, Institute of Infectious Agents, Laboratory associated with the National Reference Centre for Respiratory Infection Viruses, Hospices Civils de Lyon, IAI, North Biology Centre, North Hospital Group, 69004 Lyon, France
| | - Mary-Anne Trabaud
- Virology Laboratory, Institute of Infectious Agents, Laboratory associated with the National Reference Centre for Respiratory Infection Viruses, Hospices Civils de Lyon, IAI, North Biology Centre, North Hospital Group, 69004 Lyon, France
| | - Muriel Rabilloud
- Biostatistics Department, Hospices Civils de Lyon, Lyon France. CNRS, UMR 5558, University of Lyon, Biometrics and Evolutionary Biology Laboratory, Biostatistics-Health Team, 69100 Villeurbanne, France
| | - Amna Abichou-Klich
- Biostatistics Department, Hospices Civils de Lyon, Lyon France. CNRS, UMR 5558, University of Lyon, Biometrics and Evolutionary Biology Laboratory, Biostatistics-Health Team, 69100 Villeurbanne, France
| | - Sophie Trouillet-Assant
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France.
| |
Collapse
|
10
|
Krishna B, Metaxaki M, Perera M, Wills M, Sithole N. Comparison of different T cell assays for the retrospective determination of SARS-CoV-2 infection. J Gen Virol 2024; 105. [PMID: 39704047 DOI: 10.1099/jgv.0.002055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
It is important to be able to retrospectively determine severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections with high accuracy, both for post-coronavirus disease 2019 (COVID-19) epidemiological studies, and to distinguish between Long COVID and other multi-syndromic diseases that have overlapping symptoms. Although serum antibody levels can be measured to retrospectively diagnose SARS-CoV-2 infections, peptide stimulation of memory T cell responses is a more sensitive approach. This is because robust memory T cells are generated after SARS-CoV-2 infection and persist even after antibodies wane below detectability thresholds. In this study, we compare T cell responses using FluoroSpot-based methods and overnight stimulation of whole blood with SARS-CoV-2 peptides followed by an ELISA. Both approaches have comparable sensitivity and specificity but require different equipment and samples to be used. Furthermore, the elimination of peptides that cross-react with other coronaviruses increases the assay specificity but trades off some sensitivity. Finally, this approach can be used on archival, cryopreserved PBMCs. This work shows comparative advantages for several methods to measure SARS-CoV-2 T cell responses that could be utilized by any laboratory studying the effects of the coronavirus disease 2019 pandemic.
Collapse
Affiliation(s)
- Benjamin Krishna
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Marina Metaxaki
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Marianne Perera
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Mark Wills
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Nyarie Sithole
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| |
Collapse
|
11
|
Oliveira K, Almeida A, Silva C, Brito M, Ribeiro E. SARS-CoV-2 Immunization Index in the Academic Community: A Retrospective Post-Vaccination Study. Infect Dis Rep 2024; 16:1084-1097. [PMID: 39728010 DOI: 10.3390/idr16060088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES The COVID-19 pandemic has revolutionized vaccine production and compelled a massive global vaccination campaign. This study aimed to estimate the positivity and levels of SARS-CoV-2 IgG antibodies acquired due to vaccination and infection in the academic population of a Portuguese university. METHODS Blood samples were collected and analyzed through the ELISA methodology, and statistical analysis was performed. RESULTS A total of 529 volunteers with at least one dose of the vaccine were enrolled in this study. Individuals without a prior COVID-19 diagnosis were divided into two groups: 350, who received a full vaccination, and 114, who received a full vaccination and a booster dose of the same vaccine (81) and mixed vaccines (33). Regarding the individuals who reported a prior SARS-CoV-2 infection, 31 received a full vaccination, and 34 received only one vaccination dose. Data analysis showed a higher level of IgG against SARS-CoV-2 in individuals who were younger, female, who received the Moderna vaccine, with recent post-vaccine administration, a mixed booster dose, and prior SARS-CoV-2 infection. CONCLUSIONS Assessing vaccination's effectiveness and group immunity is crucial for pandemic management, particularly in academic environments with high individual mobility, in order to define groups at risk and redirect infection control strategies.
Collapse
Affiliation(s)
- Keltyn Oliveira
- Health & Technology Research Center, Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, Parque das Nações, 1990-096 Lisboa, Portugal
| | - Ana Almeida
- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, Parque das Nações, 1990-096 Lisboa, Portugal
| | - Carina Silva
- Health & Technology Research Center, Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, Parque das Nações, 1990-096 Lisboa, Portugal
- Centro de Estatística e Aplicações, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Miguel Brito
- Health & Technology Research Center, Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, Parque das Nações, 1990-096 Lisboa, Portugal
| | - Edna Ribeiro
- Health & Technology Research Center, Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, Parque das Nações, 1990-096 Lisboa, Portugal
| |
Collapse
|
12
|
Baldovin T, Leoni D, Geppini R, Miatton A, Amoruso I, Fonzo M, Bertoncello C, Finco M, Mazzitelli M, Sasset L, Cattelan A, Baldo V. Immunogenicity and Determinants of Antibody Response to the BNT162b2 mRNA Vaccine: A Longitudinal Study in a Cohort of People Living with HIV. Vaccines (Basel) 2024; 12:1172. [PMID: 39460338 PMCID: PMC11512344 DOI: 10.3390/vaccines12101172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic posed significant challenges worldwide, with SARS-CoV-2 vaccines critical in reducing morbidity and mortality. This study evaluates the immunogenicity and antibody persistence of the BNT162b2 vaccine in people living with HIV (PLWH). METHODS We monitored anti-SARS-CoV-2 Spike IgG concentration in a cohort of PLWH at five time points (T0-T4) using chemiluminescent microparticle immunoassays (CMIAs) at the baselined both during and after vaccination. In severely immunocompromised individuals, a boosting dose was recommended, and participants and IgG concentration were measured in the two subgroups (boosted and not boosted). RESULTS In total, 165 PLWH were included, and 83% were male with a median age of 55 years (IQR: 47-62). At T1, 161 participants (97.6%) showed seroconversion with a median of IgG values of 468.8 AU/mL (IQR: 200.4-774.3 AU/mL). By T2, all subjects maintained a positive result, with the median anti-SARS-CoV-2 Spike IgG concentration increasing to 6191.6 AU/mL (IQR: 3666.7-10,800.8 AU/mL). At T3, all participants kept their antibody levels above the positivity threshold with a median of 1694.3 AU/mL (IQR: 926.3-2966.4 AU/mL). At T4, those without a booster dose exhibited a marked decrease to a median of 649.1 AU/mL (IQR: 425.5-1299.8 AU/mL), whereas those with a booster experienced a significant increase to a median of 13,105.2 AU/mL (IQR: 9187.5-18,552.1 AU/mL). The immune response was negatively influenced by the presence of dyslipidaemia at T1 (aOR 4.75, 95% CI: 1.39-16.20) and diabetes at T3 (aOR 7.11, 95% CI: 1.10-46.1), while the use of protease inhibitors (aORs 0.06, 95% CI: 0.01-0.91) and being female (aOR 0.02, 95% CI: 0.01-0.32) at T3 were protective factors. CONCLUSIONS The immunogenicity of the BNT162b2 vaccine in PLWH has been confirmed, with booster doses necessary to maintain high levels of anti-SARS-CoV-2 Spike IgG antibodies, especially in patients with comorbidities. These findings underline the importance of a personalized vaccination strategy in this population.
Collapse
Affiliation(s)
- Tatjana Baldovin
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy (R.G.); (A.M.); (I.A.); (M.F.); (V.B.)
| | - Davide Leoni
- Infectious and Tropical Diseases Unit, Padua University Hospital, 35131 Padua, Italy; (D.L.); (M.F.); (L.S.); (A.C.)
| | - Ruggero Geppini
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy (R.G.); (A.M.); (I.A.); (M.F.); (V.B.)
| | - Andrea Miatton
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy (R.G.); (A.M.); (I.A.); (M.F.); (V.B.)
| | - Irene Amoruso
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy (R.G.); (A.M.); (I.A.); (M.F.); (V.B.)
| | - Marco Fonzo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy (R.G.); (A.M.); (I.A.); (M.F.); (V.B.)
| | - Chiara Bertoncello
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy (R.G.); (A.M.); (I.A.); (M.F.); (V.B.)
| | - Mascia Finco
- Infectious and Tropical Diseases Unit, Padua University Hospital, 35131 Padua, Italy; (D.L.); (M.F.); (L.S.); (A.C.)
| | - Maria Mazzitelli
- Infectious and Tropical Diseases Unit, Padua University Hospital, 35131 Padua, Italy; (D.L.); (M.F.); (L.S.); (A.C.)
| | - Lolita Sasset
- Infectious and Tropical Diseases Unit, Padua University Hospital, 35131 Padua, Italy; (D.L.); (M.F.); (L.S.); (A.C.)
| | - Annamaria Cattelan
- Infectious and Tropical Diseases Unit, Padua University Hospital, 35131 Padua, Italy; (D.L.); (M.F.); (L.S.); (A.C.)
- Department of Molecular Medicine, University of Padua, 35131 Padua, Italy
| | - Vincenzo Baldo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy (R.G.); (A.M.); (I.A.); (M.F.); (V.B.)
| |
Collapse
|
13
|
Alirezaylavasani A, Skeie LG, Egner IM, Chopra A, Dahl TB, Prebensen C, Vaage JT, Halvorsen B, Lund-Johansen F, Tonby K, Reikvam DH, Stiksrud B, Holter JC, Dyrhol-Riise AM, Munthe LA, Kared H. Vaccine responses and hybrid immunity in people living with HIV after SARS-CoV-2 breakthrough infections. NPJ Vaccines 2024; 9:185. [PMID: 39384763 PMCID: PMC11464709 DOI: 10.1038/s41541-024-00972-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 09/19/2024] [Indexed: 10/11/2024] Open
Abstract
The COVID-19 pandemic posed a challenge for people living with HIV (PLWH), particularly immune non-responders (INR) with compromised CD4 T-cell reconstitution following antiretroviral therapy (CD4 count <350 cells per mm3). Their diminished vaccine responses raised concerns about their vulnerability to SARS-CoV-2 breakthrough infections (BTI). Our in-depth study here revealed chronic inflammation in PLWH and a limited anti-Spike IgG response after vaccination in INR. Nevertheless, the imprinting of Spike-specific B cells by vaccination significantly enhanced the humoral responses after BTI. Notably, the magnitude of cellular CD4 response in all PLWH was comparable to that in healthy donors (HD). However, the polyfunctionality and phenotype of Spike-specific CD8 T cells in INR differed from controls. The findings highlight the need for additional boosters with variant vaccines, and for monitoring ART adherence and the durability of both humoral and cellular anti-SARS-CoV-2 immunity in INR.
Collapse
Affiliation(s)
- Amin Alirezaylavasani
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Linda Gail Skeie
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Ingrid Marie Egner
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Adity Chopra
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Tuva Børresdatter Dahl
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Christian Prebensen
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - John Torgils Vaage
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Kristian Tonby
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dag Henrik Reikvam
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Birgitte Stiksrud
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Jan Cato Holter
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Anne Ma Dyrhol-Riise
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ludvig A Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| | - Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| |
Collapse
|
14
|
Martens B, Van Caeseele P, Bullard J, Loeppky C, Wei Y, Reimer J, McKinnon LR, Shaw SY, Kindrachuk J, Stein DR. A Population-Based Study of SARS-CoV-2 IgG Antibody Responses to Vaccination in Manitoba. Vaccines (Basel) 2024; 12:1095. [PMID: 39460263 PMCID: PMC11511381 DOI: 10.3390/vaccines12101095] [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: 04/29/2024] [Revised: 07/10/2024] [Accepted: 07/22/2024] [Indexed: 10/28/2024] Open
Abstract
Understanding variables that influence antibody responses to COVID-19 vaccination within a population can provide valuable information on future vaccination strategies. In this population-based study, we examined the antibody responses to COVID-19 vaccination in Manitoba using residual serum specimens collected between January 2021 and March 2022 (n = 20,365). Samples were tested for spike and nucleocapsid IgG against SARS-CoV-2 using clinically validated assays. We assessed the impacts of multiple factors on post-vaccination antibody titres including type of vaccine, age, sex, geographic location, number of doses received, and timing of vaccination. Our investigation demonstrated that vaccination with one dose of Moderna mRNA-1273 elicited higher anti-spike IgG titres overall compared to Pfizer BNT162b2 vaccination, while one dose of Pfizer BNT162b2 followed by a second dose of Moderna mRNA-1273 exhibited higher titres than two doses of Pfizer BNT162b2 or Moderna mRNA-1273, irrespective of age. Age and time post-vaccination had considerable effects on antibody responses, with older age groups exhibiting lower anti-spike IgG titres than younger ages, and titres of those vaccinated with Pfizer BNT162b2 waning faster than those vaccinated with Moderna mRNA-1273 or a combination of Pfizer BNT162b2 and Moderna mRNA-1273. Antibody titres did not appear to be affected by sex or geographic location. Our results identify how factors such as age and type of vaccine can influence antibody responses to vaccination, and how antibody titres wane over time. This information highlights the importance of tailoring vaccine regimens to specific populations, especially those at increased risk of severe COVID-19 and can be used to inform future vaccination strategies, scheduling of booster doses, and public health measures.
Collapse
Affiliation(s)
- Brielle Martens
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Paul Van Caeseele
- Department of Pediatrics and Child Health, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3A 1S1, Canada;
- Cadham Provincial Laboratory, Winnipeg, MB R3E 3J7, Canada
| | - Jared Bullard
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Cadham Provincial Laboratory, Winnipeg, MB R3E 3J7, Canada
| | - Carla Loeppky
- Epidemiology & Surveillance, Manitoba Health, Seniors and Active Living, Winnipeg, MB R3B 3M9, Canada
| | - Yichun Wei
- Epidemiology & Surveillance, Manitoba Health, Seniors and Active Living, Winnipeg, MB R3B 3M9, Canada
| | - Joss Reimer
- Winnipeg Regional Health Authority, Winnipeg, MB R3B 1E2, Canada
| | - Lyle R. McKinnon
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa
- Department of Medical Microbiology and Immunology, University of Nairobi, Nairobi P.O. Box 19676 00202, Kenya
| | - Souradet Y. Shaw
- Department of Community Health Sciences, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Jason Kindrachuk
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
- Manitoba Centre for Proteomics and Systems Biology, Health Science Centre, Winnipeg, Manitoba R3E 3P4, Canada
| | - Derek R. Stein
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Cadham Provincial Laboratory, Winnipeg, MB R3E 3J7, Canada
| |
Collapse
|
15
|
Møller M, Friis-Hansen L, Kirkby N, Dilling-Hansen C, Andersson M, Vedsted P, Mølbak K, Koch A. Robust immune response to COVID-19 vaccination in the island population of Greenland. COMMUNICATIONS MEDICINE 2024; 4:173. [PMID: 39242878 PMCID: PMC11379896 DOI: 10.1038/s43856-024-00602-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 08/29/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND In Greenland, the COVID-19 pandemic was characterised by a late onset of community transmission and a low impact on the healthcare system, hypothesised as being partly due to a high uptake of vaccinations. To underpin this description, we aimed to assess the SARS-CoV-2 immune response post-vaccination in a Greenlandic population. METHODS In this observational cohort study, we included 430 adults in Greenland who had received a complete two-dose SARS-CoV-2 vaccination at enrolment. The total plasma SARS-CoV-2 spike glycoprotein Ig antibodies (S-Ab) induced by either the BNT162b2 or mRNA-1273 vaccine, was measured up to 11 months after the second vaccine dose. In addition, total salivary S-Abs were examined in 107 participants, and the T-cell response to the spike glycoprotein was assessed in 78 participants out of the entire study cohort. RESULTS Here we demonstrate that two months after the second vaccine dose, 96% of participants have protective plasma S-Ab levels. By 11 months, 98% have protective levels, with prior SARS-CoV-2 infection particularly enhancing S-Ab levels by 37% (95% CI 25-51%). Among individuals aged 60 years and older, we observe a 21% (95% CI 7-33%) reduction in antibody response. Total salivary S-Ab levels are detectable in all participants and significantly correlate with plasma levels. Moreover, all participants exhibit a robust SARS-CoV-2-specific T-cell response 11 months post-primary vaccination. CONCLUSIONS Our findings show that Greenlanders exhibit a robust and lasting immune response, both humoral and cellular, comparable to other population groups up to at least 11 months after the second vaccine dose. These results corroborate the hypothesis that vaccines contributed to the mild impact of the COVID-19 pandemic in the Greenlandic population.
Collapse
Affiliation(s)
- Mie Møller
- Institue of Health and Nature, University of Greenland, Nuuk, Greenland.
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark.
- Department of Internal Medicine, Queen Ingrid's Hospital, Nuuk, Greenland.
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark.
- Department of Infectious Diseases, Rigshospitalet University Hospital, Copenhagen, Denmark.
| | - Lennart Friis-Hansen
- Department of Clinical Microbiology, Rigshospitalet University Hospital, Copenhagen, Denmark
- Department of Clinical Biochemistry, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Nikolai Kirkby
- Department of Clinical Microbiology, Rigshospitalet University Hospital, Copenhagen, Denmark
| | | | - Mikael Andersson
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Vedsted
- Department of Clinical Medicine, University of Aarhus, Aarhus, Denmark
- Ilulissat Regional Hospital, Ilulissat, Greenland
| | - Kåre Mølbak
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
| | - Anders Koch
- Institue of Health and Nature, University of Greenland, Nuuk, Greenland
- Department of Internal Medicine, Queen Ingrid's Hospital, Nuuk, Greenland
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
- Department of Infectious Diseases, Rigshospitalet University Hospital, Copenhagen, Denmark
| |
Collapse
|
16
|
Reekie J, Stovring H, Nielsen H, Johansen IS, Benfield T, Wiese L, Stærke NB, Iversen K, Mustafa AB, Petersen KT, Juhl MR, Knudsen LS, Iversen MB, Andersen SD, Larsen FD, Baerends EAM, Lindvig SO, Rasmussen LD, Madsen LW, Bannister W, Jensen TO, Dietz LL, Ostrowski SR, Østergaard L, Tolstrup M, Lundgren JD, Søgaard OS. Development of antibody levels and subsequent decline in individuals with vaccine induced and hybrid immunity to SARS-CoV-2. Int J Infect Dis 2024; 146:107111. [PMID: 38801970 DOI: 10.1016/j.ijid.2024.107111] [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: 02/22/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024] Open
Abstract
OBJECTIVES This study aimed to compare antibody trajectories among individuals with SARS-CoV-2 hybrid and vaccine-induced immunity. METHODS Danish adults receiving three doses of BTN162b2 or mRNA-1237 were included prior to first vaccination (Day 0). SARS-CoV-2 anti-spike IgG levels were assessed before each vaccine dose, at Day 90, Day 180, 28 days after 3rd vaccination (Day 251), Day 365, and prior to 4th vaccination (Day 535). SARS-CoV-2 PCR results were extracted from the national microbiology database. Mixed-effect multivariable linear regression investigated the impact of hybrid-immunity (stratified into 4 groups: no hybrid immunity, PCR+ prior to 3rd dose, PCR+ after 3rd dose and before Day 365, PCR+ after Day 365) on anti-spike IgG trajectories. RESULTS A total of 4,936 individuals were included, 47% developed hybrid-immunity. Anti-spike IgG increases were observed in all groups at Day 251, with the highest levels in those PCR+ prior to 3rd dose (Geometric Mean; 535,647AU/mL vs. 374,665AU/mL with no hybrid-immunity, P<0.0001). Further increases were observed in participants who developed hybrid immunity after their 3rd dose. Anti-spike IgG levels declined from Day 251-535 in individuals without hybrid-immunity and in those who developed hybrid-immunity prior to their 3rd dose, with lower rate of decline in those with hybrid-immunity. CONCLUSION Hybrid-immunity results in higher and more durable antibody trajectories in vaccinated individuals.
Collapse
Affiliation(s)
- Joanne Reekie
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| | - Henrik Stovring
- Clinical Pharmacology, Pharmacy and Environmental Medicine, University of Southern Denmark, Odense, Denmark
| | - Henrik Nielsen
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Isik S Johansen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Thomas Benfield
- Department of Infectious Diseases, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Lothar Wiese
- Department of Medicine, Zealand University Hospital, Roskilde, Denmark
| | - Nina Breinholt Stærke
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Kasper Iversen
- Department of Cardiology and Department of Emergency Medicine, Herlev, Denmark
| | - Ahmed Basim Mustafa
- Department of Infectious Diseases, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Maria Ruwald Juhl
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
| | | | | | | | - Fredrikke Dam Larsen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Susan Olaf Lindvig
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | - Lone Wulff Madsen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark; Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Wendy Bannister
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Tomas Oestergaard Jensen
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lisa Loksø Dietz
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark
| | - Lars Østergaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jens D Lundgren
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ole Schmeltz Søgaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
17
|
Sanada T, Honda T, Kohara M. Modeling of anti-spike IgG and neutralizing antibody waning after anti-SARS-CoV-2 mRNA vaccination. Vaccine 2024; 42:126146. [PMID: 39033078 DOI: 10.1016/j.vaccine.2024.07.047] [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: 04/15/2024] [Revised: 06/07/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
At present, mRNA-based vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are being administered on a global scale. While the efficacy of mRNA vaccines has been demonstrated, several unknowns remains. For example, as the number of booster vaccinations increases, there are uncertainties regarding how long effects of a vaccine will last and how much individual variability exists. In this study, to predict the duration of vaccine efficacy, we modeled the kinetics of antibody levels for each SARS-CoV-2 vaccination dose, incorporating predictive intervals to estimate the duration of vaccine efficacy and to account for variability among individuals. A total of 3,059 serum samples from 1,346 participants were assayed to quantify IgG antibodies specific for the S1 subunit of the S protein (anti-S1 IgG) and neutralizing antibody activities against SARS-CoV-2. A power law model was used to simulate the decay of antibody titers following vaccination, and models were constructed to assess antibody level kinetics after the second, third, fourth, and fifth vaccinations. The models assumed that booster vaccinations would significantly reduce the decline in anti-S antibody and neutralizing antibody levels, resulting in levels being maintained for a longer period. No significant differences in the decay rate of antibody levels were observed among age groups, yet the peak titers of antibody levels were significantly higher in the ≤ 39 age group than in the ≥ 60 age group following the second vaccination; these differences were not observed after the third and fourth vaccinations. The modeling of antibody level kinetics after vaccination is considered to be useful for understanding the immune status of mRNA vaccine recipients.
Collapse
Affiliation(s)
- Takahiro Sanada
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tomoko Honda
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| |
Collapse
|
18
|
Yan X, Zhao X, Du Y, Wang H, Liu L, Wang Q, Liu J, Wei S. Dynamics of anti-SARS-CoV-2 IgG antibody responses following breakthrough infection and the predicted protective efficacy: A longitudinal community-based population study in China. Int J Infect Dis 2024; 145:107075. [PMID: 38697605 DOI: 10.1016/j.ijid.2024.107075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/05/2024] Open
Abstract
OBJECTIVES To assess the dynamics of the anti-SARS-CoV-2 IgG antibody levels and their efficacy against COVID-19. METHODS We conducted a longitudinal serological analysis of 852 breakthrough COVID-19 infections among the community-based population in Yichang, China. Anti-SARS-CoV-2 IgG levels were measured by chemiluminescence at approximately 3, 4, and 9 months after infection. A linear mixed model predicted IgG antibody decline over 18 months. The effectiveness of antibodies in preventing symptomatic and severe infections was determined using an existing meta-regression model. RESULTS IgG antibodies slowly declined after breakthrough infections. Initially high at around 3 months (339.44 AU/mL, IQR: 262.78-382.95 AU/mL), levels remained significant at 9 months (297.74 AU/mL, IQR: 213.22-360.62 AU/mL). The elderly (≥60 years) had lower antibody levels compared to the young (<20 years) (P < 0.001). The protective efficacy of antibodies against symptomatic and severe infections was lower in the elderly (≥60 years) (78.34% and 86.33%) compared to the young (<20 years) (96.56% and 98.75%) after 1 year. CONCLUSION The study indicated a slow decline in anti-SARS-CoV-2 IgG antibodies, maintaining considerable efficacy for over 1 year. However, lower levels in the elderly suggest reduced protective effects, underscoring the need for age-specific vaccination strategies.
Collapse
Affiliation(s)
- Xiaolong Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zhao
- Center for Disease Control and Prevention, Yichang, Hubei, China
| | - Yin Du
- Center for Disease Control and Prevention, Yichang, Hubei, China
| | - Hao Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianhua Liu
- Center for Disease Control and Prevention, Yichang, Hubei, China
| | - Sheng Wei
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
19
|
Bouros I, Hill EM, Keeling MJ, Moore S, Thompson RN. Prioritising older individuals for COVID-19 booster vaccination leads to optimal public health outcomes in a range of socio-economic settings. PLoS Comput Biol 2024; 20:e1012309. [PMID: 39116038 PMCID: PMC11309497 DOI: 10.1371/journal.pcbi.1012309] [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: 03/08/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
The rapid development of vaccines against SARS-CoV-2 altered the course of the COVID-19 pandemic. In most countries, vaccinations were initially targeted at high-risk populations, including older individuals and healthcare workers. Now, despite substantial infection- and vaccine-induced immunity in host populations worldwide, waning immunity and the emergence of novel variants continue to cause significant waves of infection and disease. Policy makers must determine how to deploy booster vaccinations, particularly when constraints in vaccine supply, delivery and cost mean that booster vaccines cannot be administered to everyone. A key question is therefore whether older individuals should again be prioritised for vaccination, or whether alternative strategies (e.g. offering booster vaccines to the individuals who have most contacts with others and therefore drive infection) can instead offer indirect protection to older individuals. Here, we use mathematical modelling to address this question, considering SARS-CoV-2 transmission in a range of countries with different socio-economic backgrounds. We show that the population structures of different countries can have a pronounced effect on the impact of booster vaccination, even when identical booster vaccination targeting strategies are adopted. However, under the assumed transmission model, prioritising older individuals for booster vaccination consistently leads to the most favourable public health outcomes in every setting considered. This remains true for a range of assumptions about booster vaccine supply and timing, and for different assumed policy objectives of booster vaccination.
Collapse
Affiliation(s)
- Ioana Bouros
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Edward M. Hill
- Mathematics Institute, University of Warwick, Coventry, United Kingdom
- Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research (SBIDER), University of Warwick, Coventry, United Kingdom
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Matt J. Keeling
- Mathematics Institute, University of Warwick, Coventry, United Kingdom
- Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research (SBIDER), University of Warwick, Coventry, United Kingdom
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Sam Moore
- Lancaster Medical School, Lancaster University, Lancaster, United Kingdom
| | - Robin N. Thompson
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Anand P, D’Andrea E, Feldman W, Wang SV, Liu J, Brill G, DiCesare E, Lin KJ. A Dynamic Prognostic Model for Identifying Vulnerable COVID-19 Patients at High Risk of Rapid Deterioration. Pharmacoepidemiol Drug Saf 2024; 33:e5872. [PMID: 39135513 PMCID: PMC11418916 DOI: 10.1002/pds.5872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 09/25/2024]
Abstract
PURPOSE We aimed to validate and, if performance was unsatisfactory, update the previously published prognostic model to predict clinical deterioration in patients hospitalized for COVID-19, using data following vaccine availability. METHODS Using electronic health records of patients ≥18 years, with laboratory-confirmed COVID-19, from a large care-delivery network in Massachusetts, USA, from March 2020 to November 2021, we tested the performance of the previously developed prediction model and updated the prediction model by incorporating data after availability of COVID-19 vaccines. We randomly divided data into development (70%) and validation (30%) cohorts. We built a model predicting worsening in a published severity scale in 24 h by LASSO regression and evaluated performance by c-statistic and Brier score. RESULTS Our study cohort consisted of 8185 patients (Development: 5730 patients [mean age: 62; 44% female] and Validation: 2455 patients [mean age: 62; 45% female]). The previously published model had suboptimal performance using data after November 2020 (N = 4973, c-statistic = 0.60. Brier score = 0.11). After retraining with the new data, the updated model included 38 predictors including 18 changing biomarkers. Patients hospitalized after Jun 1st, 2021 (when COVID-19 vaccines became widely available in Massachusetts) were younger and had fewer comorbidities than those hospitalized before. The c-statistic and Brier score were 0.77 and 0.13 in the development cohort, and 0.73 and 0.14 in the validation cohort. CONCLUSION The characteristics of patients hospitalized for COVID-19 differed substantially over time. We developed a new dynamic model for rapid progression with satisfactory performance in the validation set.
Collapse
Affiliation(s)
- Priyanka Anand
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Elvira D’Andrea
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - William Feldman
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Shirley V. Wang
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Jun Liu
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Gregory Brill
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Elyse DiCesare
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Kueiyu Joshua Lin
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School
| |
Collapse
|
21
|
Tan Z, Yang C, Lin PH, Ramadan S, Yang W, Rashidi Z, Lang S, Shafieichaharberoud F, Gao J, Pan X, Soloff N, Wu X, Bolin S, Pyeon D, Huang X. Inducing Long Lasting B Cell and T Cell Immunity Against Multiple Variants of SARS-CoV-2 Through Mutant Bacteriophage Qβ-Receptor Binding Domain Conjugate. Adv Healthc Mater 2024; 13:e2302755. [PMID: 38733291 PMCID: PMC11305917 DOI: 10.1002/adhm.202302755] [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: 08/21/2023] [Revised: 05/04/2024] [Indexed: 05/13/2024]
Abstract
More than 3 years into the global pandemic, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains a significant threat to public health. Immunities acquired from infection or current vaccines fail to provide long term protection against subsequent infections, mainly due to their fast-waning nature and the emergence of variants of concerns (VOCs) such as Omicron. To overcome these limitations, SARS-CoV-2 Spike protein receptor binding domain (RBD)-based epitopes are investigated as conjugates with a powerful carrier, the mutant bacteriophage Qβ (mQβ). The epitope design is critical to eliciting potent antibody responses with the full length RBD being superior to peptide and glycopeptide antigens. The full length RBD conjugated with mQβ activates both humoral and cellular immune systems in vivo, inducing broad spectrum, persistent, and comprehensive immune responses effective against multiple VOCs including Delta and Omicron variants, rendering it a promising vaccine candidate.
Collapse
Affiliation(s)
- Zibin Tan
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Canchai Yang
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA
| | - Po-Han Lin
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Sherif Ramadan
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Department of Chemistry, Benha University, Benha, 13518, Egypt
| | - Weizhun Yang
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
| | - Zahra Rashidi
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Shuyao Lang
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Fatemeh Shafieichaharberoud
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Jia Gao
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Xingling Pan
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Nachy Soloff
- Hatzalah of Michigan, 13650 Oak Park Blvd., Oak Park, MI, 48237, USA
| | - Xuanjun Wu
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, Shandong, 250100, China
| | - Steven Bolin
- Veterinary Diagnostic Laboratory, Michigan State University, East Lansing, MI, 48824, USA
| | - Dohun Pyeon
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
22
|
Jarlhelt I, Hansen CB, Pérez-Alós L, Weihe P, Petersen MS, Garred P. SARS-CoV-2 anti-RBD and anti-N protein responses are differentially regulated between mother-child pairs: insight from a national study cohort at the Faroe Islands. Front Immunol 2024; 15:1418678. [PMID: 39021574 PMCID: PMC11251900 DOI: 10.3389/fimmu.2024.1418678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024] Open
Abstract
Background Knowledge about SARS-CoV-2 antibody dynamics in neonates and direct comparisons with maternal antibody responses are not well established. This study aimed to characterize and directly compare the maternal and infant antibody response in a national birth cohort from the Faroe Islands. Methods The levels of immunoglobulins (Ig) targeting the receptor binding domain (RBD) of the spike protein and the nucleocapsid protein (N protein) of SARS-CoV-2 were investigated in maternal blood and umbilical cord blood from neonates. The study included 537 neonates and 565 mothers from the Faroe Islands, and follow-up samples were collected 12 months after birth. Multiple linear regression models were used to assess associations of maternal parameters with maternal and neonatal Ig levels and pregnancy outcomes. Results The finding showed that neonates acquired varying levels of SARS-CoV-2 antibodies through transplacental transfer, and the levels were significantly influenced by the mother's vaccination and infection status. The study also found that maternal vaccination and the presence of SARS-CoV-2 antibodies targeting spike RBD were associated with gestational age and APGAR scores. Furthermore, the anti-RBD and -N protein-specific antibody response dynamics during 12 months after birth exhibited differences between mothers and children. RBD and N protein responses were maintained at follow-up in the mother's cohort, while only the N protein response was maintained at follow-up in the children's cohort. Conclusion In conclusion, SARS-CoV-2-specific immune responses in newborns rely on maternal immunity, while the persistence of SARS-CoV-2-specific Igs appears to be differently regulated between mothers and children. The study provides new insights into the dynamics of SARS-CoV-2-specific immune responses in newborns and underscores the nuanced relationship between maternal factors and neonatal humoral responses.
Collapse
Affiliation(s)
- Ida Jarlhelt
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Cecilie Bo Hansen
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Pál Weihe
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Maria Skaalum Petersen
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Gravenstein S, DeVone F, Oyebanji OA, Abul Y, Cao Y, Chan PA, Halladay CW, Rudolph JL, Nugent C, Bosch J, King CL, Wilson BM, Balazs AB, White EM, Canaday DH, McConeghy KW. Durability of immunity and clinical protection in nursing home residents following bivalent SARS-CoV-2 vaccination. EBioMedicine 2024; 105:105180. [PMID: 38861869 PMCID: PMC11215210 DOI: 10.1016/j.ebiom.2024.105180] [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: 08/02/2023] [Revised: 05/12/2024] [Accepted: 05/17/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Bivalent SARS-CoV-2 vaccines were developed to counter increasing susceptibility to emerging SARS-CoV-2 variants. We evaluated the durability of immunity and protection following first bivalent vaccination among nursing home residents. METHODS We evaluated anti-spike and neutralization titers from blood in 653 community nursing home residents before and after each monovalent booster, and a bivalent vaccine. Concurrent clinical outcomes were evaluated using electronic health record data from a separate cohort of 3783 residents of Veterans Affairs (VA) nursing homes who had received at least the primary series monovalent vaccination. Using target trial emulation, we compared VA residents who did and did not receive the bivalent vaccine to measure vaccine effectiveness against infection, hospitalization, and death. FINDINGS In the community cohort, Omicron BA.5 neutralization activity rose after each monovalent and bivalent booster vaccination regardless of prior infection history. Titers declined over time but six months post-bivalent vaccination, BA.5 neutralization persisted at detectable levels in 75% of infection-naive and 98% of prior-infected individuals. In the VA nursing home cohort, bivalent vaccine added effectiveness to monovalent booster vaccination by 18.5% for infection (95% confidence interval (CI) -5.6, 34.0%), and 29.2% for hospitalization or death (95% CI -14.2, 56.2%) over five months. INTERPRETATION The level of protection declined after bivalent vaccination over a 6 month period and may open a window of added vulnerability before the next updated vaccine becomes available, suggesting a subset of nursing home residents may benefit from an additional vaccination booster. FUNDING CDC, NIH, VHA.
Collapse
Affiliation(s)
- Stefan Gravenstein
- Warren Alpert Medical School, Brown University, Providence, RI, USA; Department of Health Services, Policy & Practice, School of Public Health, Brown University, Providence, RI, USA; Center of Innovation in Long-Term Services and Supports, Veterans Administration (VA) Medical Center, Providence, RI, USA.
| | - Frank DeVone
- Center of Innovation in Long-Term Services and Supports, Veterans Administration (VA) Medical Center, Providence, RI, USA
| | | | - Yasin Abul
- Warren Alpert Medical School, Brown University, Providence, RI, USA; Center of Innovation in Long-Term Services and Supports, Veterans Administration (VA) Medical Center, Providence, RI, USA
| | - Yi Cao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Philip A Chan
- Warren Alpert Medical School, Brown University, Providence, RI, USA; Rhode Island Department of Health, Providence, RI, USA
| | - Christopher W Halladay
- Center of Innovation in Long-Term Services and Supports, Veterans Administration (VA) Medical Center, Providence, RI, USA
| | - James L Rudolph
- Warren Alpert Medical School, Brown University, Providence, RI, USA; Department of Health Services, Policy & Practice, School of Public Health, Brown University, Providence, RI, USA; Center of Innovation in Long-Term Services and Supports, Veterans Administration (VA) Medical Center, Providence, RI, USA
| | - Clare Nugent
- Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Jürgen Bosch
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | - Brigid M Wilson
- Geriatric Research Education and Clinical Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | | | - Elizabeth M White
- Department of Health Services, Policy & Practice, School of Public Health, Brown University, Providence, RI, USA
| | - David H Canaday
- Case Western Reserve University School of Medicine, Cleveland, OH, USA; Geriatric Research Education and Clinical Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA.
| | - Kevin W McConeghy
- Department of Health Services, Policy & Practice, School of Public Health, Brown University, Providence, RI, USA; Center of Innovation in Long-Term Services and Supports, Veterans Administration (VA) Medical Center, Providence, RI, USA.
| |
Collapse
|
24
|
El Khalifi M, Britton T. SIRS epidemics with individual heterogeneity of immunity waning. J Theor Biol 2024; 587:111815. [PMID: 38614211 DOI: 10.1016/j.jtbi.2024.111815] [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: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/15/2024]
Abstract
In the current paper we analyse an extended SIRS epidemic model in which immunity at the individual level wanes gradually at exponential rate, but where the waning rate may differ between individuals, for instance as an effect of differences in immune systems. The model also includes vaccination schemes aimed to reach and maintain herd immunity. We consider both the informed situation where the individual waning parameters are known, thus allowing selection of vaccinees being based on both time since last vaccination as well as on the individual waning rate, and the more likely uninformed situation where individual waning parameters are unobserved, thus only allowing vaccination schemes to depend on time since last vaccination. The optimal vaccination policies for both the informed and uniformed heterogeneous situation are derived and compared with the homogeneous waning model (meaning all individuals have the same immunity waning rate), as well as to the classic SIRS model where immunity at the individual level drops from complete immunity to complete susceptibility in one leap. It is shown that the classic SIRS model requires least vaccines, followed by the SIRS with homogeneous gradual waning, followed by the informed situation for the model with heterogeneous gradual waning. The situation requiring most vaccines for herd immunity is the most likely scenario, that immunity wanes gradually with unobserved individual heterogeneity. For parameter values chosen to mimic COVID-19 and assuming perfect initial immunity and cumulative immunity of 12 months, the classic homogeneous SIRS epidemic suggests that vaccinating individuals every 15 months is sufficient to reach and maintain herd immunity, whereas the uninformed case for exponential waning with rate heterogeneity corresponding to a coefficient of variation being 0.5, requires that individuals instead need to be vaccinated every 4.4 months.
Collapse
Affiliation(s)
- Mohamed El Khalifi
- Department of Mathematics, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, 300 50, Morocco; Department of Mathematics, Stockholm University, Stockholm, 106 91, Sweden.
| | - Tom Britton
- Department of Mathematics, Stockholm University, Stockholm, 106 91, Sweden
| |
Collapse
|
25
|
Hamm SR, Loft JA, Pérez-Alós L, Heftdal LD, Hansen CB, Møller DL, Pries-Heje MM, Hasselbalch RB, Fogh K, Hald A, Ostrowski SR, Frikke-Schmidt R, Sørensen E, Hilsted L, Bundgaard H, Garred P, Iversen K, Perch M, Sørensen SS, Rasmussen A, Sabin CA, Nielsen SD. The Impact of Time between Booster Doses on Humoral Immune Response in Solid Organ Transplant Recipients Vaccinated with BNT162b2 Vaccines. Viruses 2024; 16:860. [PMID: 38932153 PMCID: PMC11209529 DOI: 10.3390/v16060860] [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: 05/02/2024] [Revised: 05/19/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
As solid organ transplant (SOT) recipients remain at risk of severe outcomes after SARS-CoV-2 infections, vaccination continues to be an important preventive measure. In SOT recipients previously vaccinated with at least three doses of BNT162b2, we investigated humoral responses to BNT162b2 booster doses. Anti-SARS-CoV-2 receptor binding domain (RBD) immunoglobulin G (IgG) was measured using an in-house ELISA. Linear mixed models were fitted to investigate the change in the geometric mean concentration (GMC) of anti-SARS-CoV-2 RBD IgG after vaccination in participants with intervals of more or less than six months between the last two doses of vaccine. We included 107 SOT recipients vaccinated with a BNT162b2 vaccine. In participants with an interval of more than six months between the last two vaccine doses, we found a 1.34-fold change in GMC per month (95% CI 1.25-1.44), while we found a 1.09-fold change in GMC per month (95% CI 0.89-1.34) in participants with an interval of less than six months between the last two vaccine doses, resulting in a rate ratio of 0.82 (95% CI 0.66 to 1.01, p = 0.063). In conclusion, the administration of identical COVID-19 mRNA vaccine boosters within six months to SOT recipients may result in limited humoral immunogenicity of the last dose.
Collapse
Affiliation(s)
- Sebastian Rask Hamm
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Josefine Amalie Loft
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Line Dam Heftdal
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Cecilie Bo Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Dina Leth Møller
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Mia Marie Pries-Heje
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Rasmus Bo Hasselbalch
- Department of Emergency Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Cardiology, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Kamille Fogh
- Department of Emergency Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Cardiology, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Annemette Hald
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Section 2034, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Erik Sørensen
- Department of Clinical Immunology, Section 2034, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Linda Hilsted
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen University Hospital, 2200 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kasper Iversen
- Department of Emergency Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Cardiology, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Michael Perch
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Søren Schwartz Sørensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Nephrology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Allan Rasmussen
- Department of Surgical Gastroenterology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Caroline A. Sabin
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation, Institute for Global Health, UCL, Royal Free Campus, Rowland Hill St, London NW3 2PF, UK
| | - Susanne Dam Nielsen
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Surgical Gastroenterology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| |
Collapse
|
26
|
Nakamura N, Kobashi Y, Kim KS, Park H, Tani Y, Shimazu Y, Zhao T, Nishikawa Y, Omata F, Kawashima M, Yoshida M, Abe T, Saito Y, Senoo Y, Nonaka S, Takita M, Yamamoto C, Kawamura T, Sugiyama A, Nakayama A, Kaneko Y, Jeong YD, Tatematsu D, Akao M, Sato Y, Iwanami S, Fujita Y, Wakui M, Aihara K, Kodama T, Shibuya K, Iwami S, Tsubokura M. Modeling and predicting individual variation in COVID-19 vaccine-elicited antibody response in the general population. PLOS DIGITAL HEALTH 2024; 3:e0000497. [PMID: 38701055 PMCID: PMC11068210 DOI: 10.1371/journal.pdig.0000497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 02/14/2024] [Indexed: 05/05/2024]
Abstract
As we learned during the COVID-19 pandemic, vaccines are one of the most important tools in infectious disease control. To date, an unprecedentedly large volume of high-quality data on COVID-19 vaccinations have been accumulated. For preparedness in future pandemics beyond COVID-19, these valuable datasets should be analyzed to best shape an effective vaccination strategy. We are collecting longitudinal data from a community-based cohort in Fukushima, Japan, that consists of 2,407 individuals who underwent serum sampling two or three times after a two-dose vaccination with either BNT162b2 or mRNA-1273. Using the individually reconstructed time courses of the vaccine-elicited antibody response based on mathematical modeling, we first identified basic demographic and health information that contributed to the main features of the antibody dynamics, i.e., the peak, the duration, and the area under the curve. We showed that these three features of antibody dynamics were partially explained by underlying medical conditions, adverse reactions to vaccinations, and medications, consistent with the findings of previous studies. We then applied to these factors a recently proposed computational method to optimally fit an "antibody score", which resulted in an integer-based score that can be used as a basis for identifying individuals with higher or lower antibody titers from basic demographic and health information. The score can be easily calculated by individuals themselves or by medical practitioners. Although the sensitivity of this score is currently not very high, in the future, as more data become available, it has the potential to identify vulnerable populations and encourage them to get booster vaccinations. Our mathematical model can be extended to any kind of vaccination and therefore can form a basis for policy decisions regarding the distribution of booster vaccines to strengthen immunity in future pandemics.
Collapse
Affiliation(s)
- Naotoshi Nakamura
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Yurie Kobashi
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Fukushima, Japan
| | - Kwang Su Kim
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
- Department of Science System Simulation, Pukyong National University, Busan, South Korea
- Department of Mathematics, Pusan National University, Busan, South Korea
| | - Hyeongki Park
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Yuta Tani
- Medical Governance Research Institute, Tokyo, Japan
| | - Yuzo Shimazu
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tianchen Zhao
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yoshitaka Nishikawa
- Department of General Internal Medicine, Hirata Central Hospital, Fukushima, Japan
| | - Fumiya Omata
- Department of General Internal Medicine, Hirata Central Hospital, Fukushima, Japan
| | - Moe Kawashima
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | | | - Toshiki Abe
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | | | - Yuki Senoo
- Medical Governance Research Institute, Tokyo, Japan
| | - Saori Nonaka
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Morihito Takita
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Chika Yamamoto
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Takeshi Kawamura
- Proteomics Laboratory, Isotope Science Center, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Akira Sugiyama
- Proteomics Laboratory, Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Aya Nakayama
- Proteomics Laboratory, Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Yudai Kaneko
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
- Medical & Biological Laboratories Co., Ltd, Tokyo, Japan
| | - Yong Dam Jeong
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
- Department of Mathematics, Pusan National University, Busan, South Korea
| | - Daiki Tatematsu
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Marwa Akao
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Yoshitaka Sato
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoya Iwanami
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Yasuhisa Fujita
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Masatoshi Wakui
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kazuyuki Aihara
- International Research Center for Neurointelligence, The University of Tokyo Institutes for Advanced Study, The University of Tokyo, Tokyo, Japan
| | - Tatsuhiko Kodama
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kenji Shibuya
- Soma Medical Center of Vaccination for COVID-19, Fukushima, Japan
- Tokyo Foundation for Policy Research, Tokyo, Japan
| | - Shingo Iwami
- interdisciplinary Biology Laboratory (iBLab), Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya, Japan
- Institute of Mathematics for Industry, Kyushu University, Fukuoka, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Interdisciplinary Theoretical and Mathematical Sciences Program (iTHEMS), RIKEN, Saitama, Japan
- NEXT-Ganken Program, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
- Science Groove Inc., Fukuoka, Japan
| | - Masaharu Tsubokura
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Fukushima, Japan
- Medical Governance Research Institute, Tokyo, Japan
- Minamisoma Municipal General Hospital, Fukushima, Japan
| |
Collapse
|
27
|
Livieratos A, Gogos C, Akinosoglou K. Impact of Prior COVID-19 Immunization and/or Prior Infection on Immune Responses and Clinical Outcomes. Viruses 2024; 16:685. [PMID: 38793566 PMCID: PMC11125779 DOI: 10.3390/v16050685] [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: 04/01/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Cellular and humoral immunity exhibit dynamic adaptation to the mutating SARS-CoV-2 virus. It is noteworthy that immune responses differ significantly, influenced by whether a patient has received vaccination or whether there is co-occurrence of naturally acquired and vaccine-induced immunity, known as hybrid immunity. The different immune reactions, conditional on vaccination status and the viral variant involved, bear implications for inflammatory responses, patient outcomes, pathogen transmission rates, and lingering post-COVID conditions. Considering these developments, we have performed a review of recently published literature, aiming to disentangle the intricate relationships among immunological profiles, transmission, the long-term health effects post-COVID infection poses, and the resultant clinical manifestations. This investigation is directed toward understanding the variability in the longevity and potency of cellular and humoral immune responses elicited by immunization and hybrid infection.
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
|
28
|
Korosec CS, Dick DW, Moyles IR, Watmough J. SARS-CoV-2 booster vaccine dose significantly extends humoral immune response half-life beyond the primary series. Sci Rep 2024; 14:8426. [PMID: 38637521 PMCID: PMC11026522 DOI: 10.1038/s41598-024-58811-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/03/2024] [Indexed: 04/20/2024] Open
Abstract
SARS-CoV-2 lipid nanoparticle mRNA vaccines continue to be administered as the predominant prophylactic measure to reduce COVID-19 disease pathogenesis. Quantifying the kinetics of the secondary immune response from subsequent doses beyond the primary series and understanding how dose-dependent immune waning kinetics vary as a function of age, sex, and various comorbidities remains an important question. We study anti-spike IgG waning kinetics in 152 individuals who received an mRNA-based primary series (first two doses) and a subset of 137 individuals who then received an mRNA-based booster dose. We find the booster dose elicits a 71-84% increase in the median Anti-S half life over that of the primary series. We find the Anti-S half life for both primary series and booster doses decreases with age. However, we stress that although chronological age continues to be a good proxy for vaccine-induced humoral waning, immunosenescence is likely not the mechanism, rather, more likely the mechanism is related to the presence of noncommunicable diseases, which also accumulate with age, that affect immune regulation. We are able to independently reproduce recent observations that those with pre-existing asthma exhibit a stronger primary series humoral response to vaccination than compared to those that do not, and further, we find this result is sustained for the booster dose. Finally, via a single-variate Kruskal-Wallis test we find no difference between male and female humoral decay kinetics, however, a multivariate approach utilizing Least Absolute Shrinkage and Selection Operator (LASSO) regression for feature selection reveals a statistically significant (p < 1 × 10 - 3 ), albeit small, bias in favour of longer-lasting humoral immunity amongst males.
Collapse
Affiliation(s)
- Chapin S Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
| | - David W Dick
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
| | - Iain R Moyles
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada
| | - James Watmough
- Department of Mathematics and Statistics, University of New Brunswick, 3 Bailey Dr, Fredericton, E3B 5A3, NB, Canada
| |
Collapse
|
29
|
Pušnik J, Zorn J, Monzon-Posadas WO, Peters K, Osypchuk E, Blaschke S, Streeck H. Vaccination impairs de novo immune response to omicron breakthrough infection, a precondition for the original antigenic sin. Nat Commun 2024; 15:3102. [PMID: 38600072 PMCID: PMC11006949 DOI: 10.1038/s41467-024-47451-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: 11/16/2023] [Accepted: 04/02/2024] [Indexed: 04/12/2024] Open
Abstract
Several studies have suggested the imprinting of SARS-CoV-2 immunity by original immune challenge without addressing the formation of the de novo response to successive antigen exposures. As this is crucial for the development of the original antigenic sin, we assessed the immune response against the mutated epitopes of omicron SARS-CoV-2 after vaccine breakthrough. Our data demonstrate a robust humoral response in thrice-vaccinated individuals following omicron breakthrough which is a recall of vaccine-induced memory. The humoral and memory B cell responses against the altered regions of the omicron surface proteins are impaired. The T cell responses to mutated epitopes of the omicron spike protein are present due to the high cross-reactivity of vaccine-induced T cells rather than the formation of a de novo response. Our findings, therefore, underpin the speculation that the imprinting of SARS-CoV-2 immunity by vaccination may lead to the development of original antigenic sin if future variants overcome the vaccine-induced immunity.
Collapse
Affiliation(s)
- Jernej Pušnik
- Institute of Virology, University Hospital Bonn, Bonn, Germany.
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany.
| | - Jasmin Zorn
- Institute of Virology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany
| | - Werner O Monzon-Posadas
- Institute of Virology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany
- Occupational Medicine Department, University Hospital Bonn, Bonn, Germany
| | - Kathrin Peters
- Institute of Virology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany
| | - Emmanuil Osypchuk
- Institute of Virology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany
| | - Sabine Blaschke
- Emergency Department, University Medical Center Goettingen, Goettingen, Germany
| | - Hendrik Streeck
- Institute of Virology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany
| |
Collapse
|
30
|
Underwood AP, Sølund C, Jacobsen K, Binderup A, Fernandez-Antunez C, Mikkelsen LS, Inekci D, Villadsen SL, Castruita JAS, Pinholt M, Fahnøe U, Ramirez S, Brix L, Weis N, Bukh J. Neutralizing antibody and CD8 + T cell responses following BA.4/5 bivalent COVID-19 booster vaccination in adults with and without prior exposure to SARS-CoV-2. Front Immunol 2024; 15:1353353. [PMID: 38571939 PMCID: PMC10987722 DOI: 10.3389/fimmu.2024.1353353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 02/08/2024] [Indexed: 04/05/2024] Open
Abstract
As severe acute respiratory coronavirus 2 (SARS-CoV-2) variants continue to emerge, it is important to characterize immune responses against variants which can inform on protection efficacies following booster vaccination. In this study, neutralizing breadth and antigen-specific CD8+ T cell responses were analyzed in both infection-naïve and infection-experienced individuals following administration of a booster bivalent Wuhan-Hu-1+BA.4/5 Comirnaty® mRNA vaccine. Significantly higher neutralizing titers were found after this vaccination compared to the pre-third booster vaccination time point. Further, neutralizing breadth to omicron variants, including BA.1, BA.2, BA.5, BQ.1 and XBB.1, was found to be boosted following bivalent vaccination. SARS-CoV-2-specific CD8+ T cells were identified, but with no evidence that frequencies were increased following booster vaccinations. Spike protein-specific CD8+ T cells were the only responses detected after vaccination and non-spike-specific CD8+ T cells were only detected after infection. Both spike-specific and non-spike-specific CD8+ T cells were found at much lower frequencies than CD8+ T cells specific to cytomegalovirus (CMV), Epstein-Barr virus (EBV) and influenza (Flu). Taken together, these results show that the bivalent Wuhan-Hu-1+BA.4/5 Comirnaty® mRNA vaccine boosted the breadth of neutralization to newer SARS-CoV-2 variants and that vaccination is able to induce spike protein-specific CD8+ T cell responses, which are maintained longitudinally.
Collapse
Affiliation(s)
- Alexander P. Underwood
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Christina Sølund
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | | | - Alekxander Binderup
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Carlota Fernandez-Antunez
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Lotte S. Mikkelsen
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | | | - Signe Lysemose Villadsen
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Jose A. S. Castruita
- Department of Clinical Microbiology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Mette Pinholt
- Department of Clinical Microbiology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Ulrik Fahnøe
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Santseharay Ramirez
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | | | - Nina Weis
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| |
Collapse
|
31
|
Takefuji Y. Vaccine effects on COVID-19 infection with bivalent boosting by age group. Drug Resist Updat 2024; 73:101039. [PMID: 38169273 DOI: 10.1016/j.drup.2023.101039] [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: 02/07/2023] [Revised: 08/17/2023] [Accepted: 12/24/2023] [Indexed: 01/05/2024]
Abstract
This paper examines time-series vaccine effectiveness on COVID-19 infection with/without a bivalent booster dose by 6 age groups such as 18-29, 30-49, 50-64, 65-79, 80+, and all_ages respectively. CDC's COVID data on rates of COVID-19 cases and deaths by updated (bivalent) booster status was used in this study. This result concludes that there is no difference between vaccines with or without a bivalent booster dose for preventing COVID-19 infection in 6 age groups 18-29, 30-49, 50-64, 65-79, 80+, and all_ages. Vaccination is effective in two age groups of 65-79 and 80+ for preventing COVID-19 infection. However, vaccine effectiveness against COVID-19 infection has not been confirmed in the 18-29 and 30-49 age groups.
Collapse
Affiliation(s)
- Yoshiyasu Takefuji
- Faculty of Data Science, Musashino University, 3-3-3 Ariake Koto-ku, Tokyo 135-8181, Japan.
| |
Collapse
|
32
|
Tai CG, Haviland MJ, Kissler SM, Lucia RM, Merson M, Maragakis LL, Ho DD, Anderson DJ, DiFiori J, Grubaugh ND, Grad YH, Mack CD. Low antibody levels associated with significantly increased rate of SARS-CoV-2 infection in a highly vaccinated population from the US National Basketball Association. J Med Virol 2024; 96:e29505. [PMID: 38465748 DOI: 10.1002/jmv.29505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/12/2024] [Accepted: 02/23/2024] [Indexed: 03/12/2024]
Abstract
SARS-CoV-2 antibody levels may serve as a correlate for immunity and could inform optimal booster timing. The relationship between antibody levels and protection from infection was evaluated in vaccinated individuals from the US National Basketball Association who had antibody levels measured at a single time point from September 12, 2021, to December 31, 2021. Cox proportional hazards models were used to estimate the risk of infection within 90 days of serologic testing by antibody level (<250, 250-800, and >800 AU/mL1 ), adjusting for age, time since last vaccine dose, and history of SARS-CoV-2 infection. Individuals were censored on date of booster receipt. The analytic cohort comprised 2323 individuals and was 78.2% male, 68.1% aged ≤40 years, and 56.4% vaccinated (primary series) with the Pfizer-BioNTech mRNA vaccine. Among the 2248 (96.8%) individuals not yet boosted at antibody testing, 77% completed their primary vaccine series 4-6 months before testing and the median (interquartile range) antibody level was 293.5 (interquartile range: 121.0-740.5) AU/mL. Those with levels <250 AU/mL (adj hazard ratio [HR]: 2.4; 95% confidence interval [CI]: 1.5-3.7) and 250-800 AU/mL (adj HR: 1.5; 95% CI: 0.98-2.4) had greater infection risk compared to those with levels >800 AU/mL. Antibody levels could inform individual COVID-19 risk and booster scheduling.
Collapse
Affiliation(s)
| | | | - Steven M Kissler
- Harvard University T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | - Michael Merson
- Duke University Duke Global Health Institute, Durham, North Carolina, USA
| | - Lisa L Maragakis
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Deverick J Anderson
- Duke University Center for Antimicrobial Stewardship and Infection Prevention, Durham, North Carolina, USA
| | - John DiFiori
- National Basketball Association, New York, New York, USA
- Hospital for Special Surgery, New York, New York, USA
| | - Nathan D Grubaugh
- Yale University School of Public Health, New Haven, Connecticut, USA
| | - Yonatan H Grad
- Harvard University T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | | |
Collapse
|
33
|
Kim JS, Sun Y, Balte P, Cushman M, Boyle R, Tracy RP, Styer LM, Bell TD, Anderson MR, Allen NB, Schreiner PJ, Bowler RP, Schwartz DA, Lee JS, Xanthakis V, Doyle MF, Regan EA, Make BJ, Kanaya AM, Wenzel SE, Coresh J, Isasi CR, Raffield LM, Elkind MSV, Howard VJ, Ortega VE, Woodruff P, Cole SA, Henderson JM, Mantis NJ, Parker MM, Demmer RT, Oelsner EC. Demographic and Clinical Factors Associated With SARS-CoV-2 Spike 1 Antibody Response Among Vaccinated US Adults: the C4R Study. Nat Commun 2024; 15:1492. [PMID: 38374032 PMCID: PMC10876680 DOI: 10.1038/s41467-024-45468-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 01/24/2024] [Indexed: 02/21/2024] Open
Abstract
This study investigates correlates of anti-S1 antibody response following COVID-19 vaccination in a U.S. population-based meta-cohort of adults participating in longstanding NIH-funded cohort studies. Anti-S1 antibodies were measured from dried blood spots collected between February 2021-August 2022 using Luminex-based microsphere immunoassays. Of 6245 participants, mean age was 73 years (range, 21-100), 58% were female, and 76% were non-Hispanic White. Nearly 52% of participants received the BNT162b2 vaccine and 48% received the mRNA-1273 vaccine. Lower anti-S1 antibody levels are associated with age of 65 years or older, male sex, higher body mass index, smoking, diabetes, COPD and receipt of BNT16b2 vaccine (vs mRNA-1273). Participants with a prior infection, particularly those with a history of hospitalized illness, have higher anti-S1 antibody levels. These results suggest that adults with certain socio-demographic and clinical characteristics may have less robust antibody responses to COVID-19 vaccination and could be prioritized for more frequent re-vaccination.
Collapse
Affiliation(s)
- John S Kim
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Yifei Sun
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Pallavi Balte
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Mary Cushman
- Department of Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | - Rebekah Boyle
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | - Russell P Tracy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | - Linda M Styer
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Taison D Bell
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | | | - Norrina B Allen
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Pamela J Schreiner
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Russell P Bowler
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - David A Schwartz
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Joyce S Lee
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Vanessa Xanthakis
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Margaret F Doyle
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | | | - Barry J Make
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - Alka M Kanaya
- Division of General Internal Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Sally E Wenzel
- Department of Medicine, Department of Immunology, and Department of Environmental Medicine and Occupational Health, University of Pittsburgh School of Medicine, School of Public Health, Pittsburgh, PA, USA
| | - Josef Coresh
- Department of Population Health, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA
- Department of Medicine, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Carmen R Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Mitchell S V Elkind
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Virginia J Howard
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victor E Ortega
- Division of Respiratory Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Prescott Woodruff
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Shelley A Cole
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Joel M Henderson
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Nicholas J Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, USA
| | - Monica M Parker
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA.
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA.
- Division of Epidemiology, Department of Quantitative Health Sciences, College of Medicine and Science, Mayo Clinic, Rochester, MN, USA.
| | - Elizabeth C Oelsner
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
34
|
Tsurkalenko O, Bulaev D, O'Sullivan MP, Snoeck C, Ghosh S, Kolodkin A, Rommes B, Gawron P, Moreno CV, Gomes CPC, Kaysen A, Ohnmacht J, Schröder VE, Pavelka L, Meyers GR, Pauly L, Pauly C, Hanff AM, Meyrath M, Leist A, Sandt E, Aguayo GA, Perquin M, Gantenbein M, Abdelrahman T, Klucken J, Satagopam V, Hilger C, Turner J, Vaillant M, Fritz JV, Ollert M, Krüger R. Creation of a pandemic memory by tracing COVID-19 infections and immunity in Luxembourg (CON-VINCE). BMC Infect Dis 2024; 24:179. [PMID: 38336649 PMCID: PMC10858600 DOI: 10.1186/s12879-024-09055-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND During the COVID-19 pandemic swift implementation of research cohorts was key. While many studies focused exclusively on infected individuals, population based cohorts are essential for the follow-up of SARS-CoV-2 impact on public health. Here we present the CON-VINCE cohort, estimate the point and period prevalence of the SARS-CoV-2 infection, reflect on the spread within the Luxembourgish population, examine immune responses to SARS-CoV-2 infection and vaccination, and ascertain the impact of the pandemic on population psychological wellbeing at a nationwide level. METHODS A representative sample of the adult Luxembourgish population was enrolled. The cohort was followed-up for twelve months. SARS-CoV-2 RT-qPCR and serology were conducted at each sampling visit. The surveys included detailed epidemiological, clinical, socio-economic, and psychological data. RESULTS One thousand eight hundred sixty-five individuals were followed over seven visits (April 2020-June 2021) with the final weighted period prevalence of SARS-CoV-2 infection of 15%. The participants had similar risks of being infected regardless of their gender, age, employment status and education level. Vaccination increased the chances of IgG-S positivity in infected individuals. Depression, anxiety, loneliness and stress levels increased at a point of study when there were strict containment measures, returning to baseline afterwards. CONCLUSION The data collected in CON-VINCE study allowed obtaining insights into the infection spread in Luxembourg, immunity build-up and the impact of the pandemic on psychological wellbeing of the population. Moreover, the study holds great translational potential, as samples stored at the biobank, together with self-reported questionnaire information, can be exploited in further research. TRIAL REGISTRATION Trial registration number: NCT04379297, 10 April 2020.
Collapse
Affiliation(s)
| | - Dmitry Bulaev
- Luxembourg Institute of Health, Strassen, Luxembourg
| | | | | | | | | | | | - Piotr Gawron
- University of Luxembourg, Esch-Belval, Luxembourg
| | | | | | - Anne Kaysen
- University of Luxembourg, Esch-Belval, Luxembourg
| | | | - Valerie E Schröder
- University of Luxembourg, Esch-Belval, Luxembourg
- Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Lukas Pavelka
- Luxembourg Institute of Health, Strassen, Luxembourg
- Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Guilherme Ramos Meyers
- Luxembourg Institute of Health, Strassen, Luxembourg
- University of Luxembourg, Esch-Belval, Luxembourg
| | - Laure Pauly
- Luxembourg Institute of Health, Strassen, Luxembourg
| | - Claire Pauly
- Luxembourg Institute of Health, Strassen, Luxembourg
- Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Anne-Marie Hanff
- Luxembourg Institute of Health, Strassen, Luxembourg
- University of Luxembourg, Esch-Belval, Luxembourg
- Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Max Meyrath
- Luxembourg Institute of Health, Strassen, Luxembourg
| | - Anja Leist
- University of Luxembourg, Esch-Belval, Luxembourg
| | - Estelle Sandt
- Luxembourg Institute of Health, Strassen, Luxembourg
| | | | | | | | | | - Jochen Klucken
- Luxembourg Institute of Health, Strassen, Luxembourg
- University of Luxembourg, Esch-Belval, Luxembourg
| | | | | | | | | | | | - Markus Ollert
- Luxembourg Institute of Health, Strassen, Luxembourg
| | - Rejko Krüger
- Luxembourg Institute of Health, Strassen, Luxembourg
- University of Luxembourg, Esch-Belval, Luxembourg
- Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| |
Collapse
|
35
|
Moore R, Purvis RS, Willis DE, Li J, Langner J, Gurel-Headley M, Kraleti S, Curran GM, Macechko MD, McElfish PA. "Every Time It Comes Time for Another Shot, It's a Re-Evaluation": A Qualitative Study of Intent to Receive COVID-19 Boosters among Parents Who Were Hesitant Adopters of the COVID-19 Vaccine. Vaccines (Basel) 2024; 12:171. [PMID: 38400154 PMCID: PMC10892107 DOI: 10.3390/vaccines12020171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
COVID-19 vaccine coverage remains low for US children, especially among those living in rural areas and the Southern/Southeastern US. As of 12 September 2023, the CDC recommended bivalent booster doses for everyone 6 months and older. Emerging research has shown an individual may be vaccine hesitant and also choose to receive a vaccine for themselves or their child(ren); however, little is known regarding how hesitant adopters evaluate COVID-19 booster vaccinations. We used an exploratory qualitative descriptive study design and conducted individual interviews with COVID-19 vaccine-hesitant adopter parents (n = 20) to explore COVID-19 parental intentions to have children receive COVID-19 boosters. Three primary themes emerged during the analysis: risk, confidence, and intent, with risk assessments from COVID-19 and COVID-19 vaccine confidence often related to an individual parent's intent to vaccinate. We also found links among individuals with persistent concerns about the COVID-19 vaccine and low COVID-19 vaccine confidence with conditional and/or low/no intent and refusal to receive recommended boosters for children. Our findings suggest that healthcare providers and public health officials should continue making strong recommendations for vaccines, continue to address parental concerns, and provide strong evidence for vaccine safety and efficacy even among the vaccinated.
Collapse
Affiliation(s)
- Ramey Moore
- College of Medicine, University of Arkansas for Medical Sciences Northwest, 2708 S. 48th St., Springdale, AR 72762, USA; (R.M.); (R.S.P.); (D.E.W.)
| | - Rachel S. Purvis
- College of Medicine, University of Arkansas for Medical Sciences Northwest, 2708 S. 48th St., Springdale, AR 72762, USA; (R.M.); (R.S.P.); (D.E.W.)
| | - Don E. Willis
- College of Medicine, University of Arkansas for Medical Sciences Northwest, 2708 S. 48th St., Springdale, AR 72762, USA; (R.M.); (R.S.P.); (D.E.W.)
| | - Ji Li
- Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences Northwest, 2708 S. 48th St., Springdale, AR 72762, USA
| | - Jonathan Langner
- Office of Community Health and Research, University of Arkansas for Medical Sciences Northwest, 2708 S. 48th St., Springdale, AR 72762, USA;
| | - Morgan Gurel-Headley
- College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock, AR 72205, USA; (M.G.-H.); (S.K.)
- Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock, AR 72205, USA
| | - Shashank Kraleti
- College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock, AR 72205, USA; (M.G.-H.); (S.K.)
| | - Geoffrey M. Curran
- College of Pharmacy, University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock, AR 72205, USA;
- Center for Mental Healthcare and Outcomes Research, Central Arkansas Veterans Healthcare System, 4300 W. 7th St., North Little Rock, AR 72114, USA
| | - Michael D. Macechko
- College of Medicine, University of Arkansas for Medical Sciences Northwest, 1125 N. College Ave., Fayetteville, AR 72703, USA;
| | - Pearl A. McElfish
- College of Medicine, University of Arkansas for Medical Sciences Northwest, 2708 S. 48th St., Springdale, AR 72762, USA; (R.M.); (R.S.P.); (D.E.W.)
| |
Collapse
|
36
|
Samanta S, Banerjee J, Das A, Das S, Ahmed R, Das S, Pal A, Ali KM, Mukhopadhyay R, Giri B, Dash SK. Enhancing Immunological Memory: Unveiling Booster Doses to Bolster Vaccine Efficacy Against Evolving SARS-CoV-2 Mutant Variants. Curr Microbiol 2024; 81:91. [PMID: 38311669 DOI: 10.1007/s00284-023-03597-2] [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: 01/09/2023] [Accepted: 12/19/2023] [Indexed: 02/06/2024]
Abstract
A growing number of re-infections with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in previously immunized individuals has sparked discussions about the potential need for a booster vaccine dosage to counteract declining antibody levels and new strains. The protective immunity produced by vaccinations, and past illnesses relies on immunological memory. CD4 + T cells, CD8 + T cells, B cells, and long-lasting antibody responses are all components of the adaptive immune system that can generate and maintain this immunological memory. Since novel mutant variants have emerged one after the other, the world has been hit by repeated waves. Various vaccine formulations against SARS-CoV-2 have been administered across the globe. Thus, estimating the efficacy of those vaccines against gradually developed mutant stains is the essential parameter regarding the fate of those vaccine formulations and the necessity of booster doses and their frequency. In this review, focus has also been given to how vaccination stacks up against moderate and severe acute infections in terms of the longevity of the immune cells, neutralizing antibody responses, etc. However, hybrid immunity shows a greater accuracy of re-infection of variants of concern (VOCs) of SARS-CoV-2 than infection and immunization. The review conveys knowledge of detailed information about several marketed vaccines and the status of their efficacy against specific mutant strains of SARS-CoV-2. Furthermore, this review discusses the status of immunological memory after infection, mixed infection, and vaccination.
Collapse
Affiliation(s)
- Sovan Samanta
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Jhimli Banerjee
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Aparna Das
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Sourav Das
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Rubai Ahmed
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Swarnali Das
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Amitava Pal
- Department of Physiology, City College, 102/1, Raja Rammohan Sarani, Kolkata, 700009, West Bengal, India
| | - Kazi Monjur Ali
- Department of Nutrition, Maharajadhiraj Uday Chand Women's College, B.C. Road, Bardhaman, 713104, West Bengal, India
| | - Rupanjan Mukhopadhyay
- Department of Physiology, City College, 102/1, Raja Rammohan Sarani, Kolkata, 700009, West Bengal, India
| | - Biplab Giri
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Sandeep Kumar Dash
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India.
| |
Collapse
|
37
|
Höft MA, Burgers WA, Riou C. The immune response to SARS-CoV-2 in people with HIV. Cell Mol Immunol 2024; 21:184-196. [PMID: 37821620 PMCID: PMC10806256 DOI: 10.1038/s41423-023-01087-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 10/13/2023] Open
Abstract
This review examines the intersection of the HIV and SARS-CoV-2 pandemics. People with HIV (PWH) are a heterogeneous group that differ in their degree of immune suppression, immune reconstitution, and viral control. While COVID-19 in those with well-controlled HIV infection poses no greater risk than that for HIV-uninfected individuals, people with advanced HIV disease are more vulnerable to poor COVID-19 outcomes. COVID-19 vaccines are effective and well tolerated in the majority of PWH, though reduced vaccine efficacy, breakthrough infections and faster waning of vaccine effectiveness have been demonstrated in PWH. This is likely a result of suboptimal humoral and cellular immune responses after vaccination. People with advanced HIV may also experience prolonged infection that may give rise to new epidemiologically significant variants, but initiation or resumption of antiretroviral therapy (ART) can effectively clear persistent infection. COVID-19 vaccine guidelines reflect these increased risks and recommend prioritization for vaccination and additional booster doses for PWH who are moderately to severely immunocompromised. We recommend continued research and monitoring of PWH with SARS-CoV-2 infection, especially in areas with a high HIV burden.
Collapse
Affiliation(s)
- Maxine A Höft
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Wendy A Burgers
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa.
| | - Catherine Riou
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
38
|
Serwanga J, Ankunda V, Katende JS, Baine C, Oluka GK, Odoch G, Nantambi H, Mugaba S, Namuyanja A, Ssali I, Ejou P, Kato L, Musenero M, Kaleebu P. Sustained S-IgG and S-IgA antibodies to Moderna's mRNA-1273 vaccine in a Sub-Saharan African cohort suggests need for booster timing reconsiderations. Front Immunol 2024; 15:1348905. [PMID: 38357547 PMCID: PMC10864610 DOI: 10.3389/fimmu.2024.1348905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024] Open
Abstract
Introduction This study sought to elucidate the long-term antibody responses to the Moderna mRNA-1273 COVID-19 vaccine within a Ugandan cohort, aiming to contribute to the sparse data on m-RNA vaccine immunogenicity in Sub-Saharan Africa. Methods We tracked the development and persistence of the elicited antibodies in 19 participants aged 18 to 67, who received two doses of the mRNA-1273 vaccine. A validated enzyme-linked immunosorbent assay (ELISA) was used to quantify SARS-CoV-2-specific IgG, IgM, and IgA antibodies against the spike (S) and nucleoproteins (N). The study's temporal scope extended from the baseline to one year, capturing immediate and long-term immune responses. Statistical analyses were performed using the Wilcoxon test to evaluate changes in antibody levels across predetermined intervals with the Hochberg correction for multiple comparisons. Results Our results showed a significant initial rise in spike-directed IgG (S-IgG) and spike-directed IgA (S-IgA) levels, which remained elevated for the duration of the study. The S-IgG concentrations peaked 14 days afterboosting, while spike-directed IgM (S-IgM) levels were transient, aligning with their early response role. Notably, post-booster antibody concentrations did not significantly change. Prior S-IgG status influenced the post-priming S-IgA dynamics, with baseline S-IgG positive individuals maintaining higher S-IgA responses, a difference that did not reach statistical difference post-boost. Three instances of breakthrough infections: two among participants who exhibited baseline seropositivity for S-IgG, and one in a participant initially seronegative for S-IgG. Discussion In conclusion, the mRNA-1273 vaccine elicited robust and persistent S-IgG and S-IgA antibody responses, particularly after the first dose, indicating potential for long-term immunity. Prior viral exposure enhances post-vaccination S-IgA responses compared to naive individuals, which aligned with the prior-naïve, post-boost. The stable antibody levels observed post-booster dose, remaining high over an extended period, with no significant secondary rise, and no difference by baseline exposure, suggest that initial vaccination may sufficiently prime the immune system for prolonged protection in this population, allowing for potential to delay booster schedules as antibody responses remained high at the time of boosting. This finding calls for a reassessment of the booster dose scheduling in this demographic.
Collapse
Affiliation(s)
- Jennifer Serwanga
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Violet Ankunda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Joseph Ssebwana Katende
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Claire Baine
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Gerald Kevin Oluka
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Geoffrey Odoch
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Hellen Nantambi
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Susan Mugaba
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Angella Namuyanja
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Ivan Ssali
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Peter Ejou
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Laban Kato
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Monica Musenero
- Science, Technology, and Innovation Secretariat, Office of the President, Government of Uganda, Kampala, Uganda
| | - Pontiano Kaleebu
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| |
Collapse
|
39
|
Ghildiyal T, Rai N, Mishra Rawat J, Singh M, Anand J, Pant G, Kumar G, Shidiki A. Challenges in Emerging Vaccines and Future Promising Candidates against SARS-CoV-2 Variants. J Immunol Res 2024; 2024:9125398. [PMID: 38304142 PMCID: PMC10834093 DOI: 10.1155/2024/9125398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/10/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024] Open
Abstract
Since the COVID-19 outbreak, the severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) virus has evolved into variants with varied infectivity. Vaccines developed against COVID-19 infection have boosted immunity, but there is still uncertainty on how long the immunity from natural infection or vaccination will last. The present study attempts to outline the present level of information about the contagiousness and spread of SARS-CoV-2 variants of interest and variants of concern (VOCs). The keywords like COVID-19 vaccine types, VOCs, universal vaccines, bivalent, and other relevant terms were searched in NCBI, Science Direct, and WHO databases to review the published literature. The review provides an integrative discussion on the current state of knowledge on the type of vaccines developed against SARS-CoV-2, the safety and efficacy of COVID-19 vaccines concerning the VOCs, and prospects of novel universal, chimeric, and bivalent mRNA vaccines efficacy to fend off existing variants and other emerging coronaviruses. Genomic variation can be quite significant, as seen by the notable differences in impact, transmission rate, morbidity, and death during several human coronavirus outbreaks. Therefore, understanding the amount and characteristics of coronavirus genetic diversity in historical and contemporary strains can help researchers get an edge over upcoming variants.
Collapse
Affiliation(s)
- Tanmay Ghildiyal
- Department of Microbial Biotechnology, Panjab University, Chandigarh, India
| | - Nishant Rai
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, India
| | - Janhvi Mishra Rawat
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, India
| | - Maargavi Singh
- Department of Instrumentation and Control Engineering, Manipal Institute of Technology, Manipal, Karnataka, India
| | - Jigisha Anand
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, India
| | - Gaurav Pant
- Department of Microbiology, Graphic Era Deemed to be University, Dehradun, India
| | - Gaurav Kumar
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India
| | | |
Collapse
|
40
|
Kameda T, Utsunomiya A, Otsuka N, Kubuki Y, Uchida T, Shide K, Kamiunten A, Nakano N, Tokunaga M, Miyazono T, Ito Y, Yonekura K, Kawakita T, Akizuki K, Tahira Y, Karasawa M, Hidaka T, Konagata A, Taniguchi N, Nagatomo Y, Kogo F, Shimizu K, Ueno H, Ishizaki J, Takahashi N, Ikei Y, Hidaka M, Yamaguchi H, Shimoda K. Impaired humoral immunity following COVID-19 vaccination in HTLV-1 carriers. BMC Infect Dis 2024; 24:96. [PMID: 38233756 PMCID: PMC10792913 DOI: 10.1186/s12879-024-09001-z] [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: 08/28/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Whether human T-lymphotropic virus type 1 (HTLV-1) carriers can develop sufficient humoral immunity after coronavirus disease 2019 (COVID-19) vaccination is unknown. METHODS To investigate humoral immunity after COVID-19 vaccination in HTLV-1 carriers, a multicenter, prospective observational cohort study was conducted at five institutions in southwestern Japan, an endemic area for HTLV-1. HTLV-1 carriers and HTLV-1-negative controls were enrolled for this study from January to December 2022. During this period, the third dose of the COVID-19 vaccine was actively administered. HTLV-1 carriers were enrolled during outpatient visits, while HTLV-1-negative controls included health care workers and patients treated by participating institutions for diabetes, hypertension, or dyslipidemia. The main outcome was the effect of HTLV-1 infection on the plasma anti-COVID-19 spike IgG (IgG-S) titers after the third dose, assessed by multivariate linear regression with other clinical factors. RESULTS We analyzed 181 cases (90 HTLV-1 carriers, 91 HTLV-1-negative controls) after receiving the third dose. HTLV-1 carriers were older (median age 67.0 vs. 45.0 years, p < 0.001) and more frequently had diabetes, hypertension, or dyslipidemia than did HTLV-1-negative controls (60.0% vs. 27.5%, p < 0.001). After the third dose, the IgG-S titers decreased over time in both carriers and controls. Multivariate linear regression in the entire cohort showed that time since the third dose, age, and HTLV-1 infection negatively influenced IgG-S titers. After adjusting for confounders such as age, or presence of diabetes, hypertension, or dyslipidemia between carriers and controls using the overlap weighting propensity score method, and performing weighted regression analysis in the entire cohort, both time since the third dose and HTLV-1 infection negatively influenced IgG-S titers. CONCLUSIONS The humoral immunity after the third vaccination dose is impaired in HTLV-1 carriers; thus, customized vaccination schedules may be necessary for them.
Collapse
Affiliation(s)
- Takuro Kameda
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Atae Utsunomiya
- Department of Hematology, Imamura General Hospital, Kagoshima, Japan
| | | | - Yoko Kubuki
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Taisuke Uchida
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Kotaro Shide
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Ayako Kamiunten
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Nobuaki Nakano
- Department of Hematology, Imamura General Hospital, Kagoshima, Japan
| | - Masahito Tokunaga
- Department of Hematology, Imamura General Hospital, Kagoshima, Japan
| | | | - Yoshikiyo Ito
- Department of Hematology, Imamura General Hospital, Kagoshima, Japan
| | - Kentaro Yonekura
- Department of Dermatology, Imamura General Hospital, Kagoshima, Japan
| | - Toshiro Kawakita
- National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Keiichi Akizuki
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Yuki Tahira
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Masayoshi Karasawa
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Tomonori Hidaka
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Ayaka Konagata
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Norifumi Taniguchi
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Yuma Nagatomo
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Fumiko Kogo
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Koichiro Shimizu
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Hiroaki Ueno
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Junzo Ishizaki
- Department of Internal Medicine, Aisenkai Nichinan Hospital, Nichinan, Japan
| | | | | | - Michihiro Hidaka
- National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Hideki Yamaguchi
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Kazuya Shimoda
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan.
| |
Collapse
|
41
|
Kim YK, Choi Y, Jung JI, Kim JY, Kim MH, Curtis J, Lee EB. Modeling of antibody responses to COVID-19 vaccination in patients with rheumatoid arthritis. Sci Rep 2024; 14:1335. [PMID: 38228736 PMCID: PMC10791674 DOI: 10.1038/s41598-024-51535-4] [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: 08/08/2023] [Accepted: 01/06/2024] [Indexed: 01/18/2024] Open
Abstract
To construct a model of the antibody response to COVID-19 vaccination in patients with rheumatoid arthritis (RA), and to identify clinical factors affecting the antibody response. A total of 779 serum samples were obtained from 550 COVID-19-naïve RA patients who were vaccinated against COVID-19. Antibody titers for the receptor binding domain (anti-RBD) and nucleocapsid (anti-N) were measured. The time from vaccination, and the log-transformed anti-RBD titer, were modeled using a fractional polynomial method. Clinical factors affecting antibody responses were analyzed by a regression model using generalized estimating equation. The anti-RBD titer peaked at about 2 weeks post-vaccination and decreased exponentially to 36.5% of the peak value after 2 months. Compared with the first vaccination, the 3rd or 4th vaccinations shifted the peaks of anti-RBD antibody response curves significantly upward (by 28-fold [4-195] and 32-fold [4-234], respectively). However, there was no significant shift in the peak from the 3rd vaccination to the 4th vaccination (p = 0.64). Multivariable analysis showed that sulfasalazine increased the vaccine response (by 1.49-fold [1.13-1.97]), but abatacept or JAK inhibitor decreased the vaccine response (by 0.13-fold [0.04-0.43] and 0.44-fold [0.26-0.74], respectively). Age was associated with lower ln [anti-RBD] values (coefficient: - 0.03 [- 0.04 to - 0.02]). In conclusion, the anti-RBD response of RA patients peaked at 2 weeks after COVID-19 vaccination, and then decreased exponentially, with the maximum peak increase observed after the 3rd vaccination. The antibody response was affected by age and the medications used to treat RA.
Collapse
Affiliation(s)
- Yun Kyu Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yunhee Choi
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ji In Jung
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ju Yeon Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Mi Hyeon Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jeffrey Curtis
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, USA
| | - Eun Bong Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 110-744, Republic of Korea.
| |
Collapse
|
42
|
Ratishvili T, Quach HQ, Haralambieva IH, Suryawanshi YR, Ovsyannikova IG, Kennedy RB, Poland GA. A multifaceted approach for identification, validation, and immunogenicity of naturally processed and in silico-predicted highly conserved SARS-CoV-2 peptides. Vaccine 2024; 42:162-174. [PMID: 38105139 DOI: 10.1016/j.vaccine.2023.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/19/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
SARS-CoV-2 remains a major global public health concern. Antibody waning and immune escape variant emergence necessitate the development of next generation vaccines that induce cross-reactive durable immune responses. T cell responses to SARS-CoV-2 demonstrate higher conservation, antigenic breadth, and longevity than antibody responses. Therefore, we sought to identify pathogen-derived T cell epitopes for a potential peptide-based vaccine. We pursued an approach leveraging: 1) liquid chromatography and tandem mass spectrometry (LC-MS/MS)-based identification of peptides from ancestral SARS-CoV-2-infected cell lines, 2) epitope prediction algorithms, and 3) overlapping peptide libraries. From this strategy, we identified 380 unique SARS-CoV-2-derived peptide sequences, including 53 antigenic HLA class I and class II peptides from multiple structural and non-structural/accessory viral proteins. These peptide sequences were highly conserved across variants of concern/interest (VoC/VoIs), and are estimated to achieve coverage of >96% of the world population. Our findings validate this discovery pipeline for peptide identification and immunogenicity testing, and are a crucial step toward the development of a next-generation multi-epitope SARS-CoV-2 peptide vaccine, and a novel vaccine platform methodology.
Collapse
Affiliation(s)
- Tamar Ratishvili
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Huy Quang Quach
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Iana H Haralambieva
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Yogesh R Suryawanshi
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Inna G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Gregory A Poland
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
43
|
Rasche MM, Kaufmann EC, Ratishvili T, Swanson IM, Ovsyannikova IG, Kennedy RB. Detection of SARS-CoV-2-Specific Cells Utilizing Whole Proteins and/or Peptides in Human PBMCs Using IFN-ƴ ELISPOT Assay. Methods Mol Biol 2024; 2768:117-133. [PMID: 38502391 DOI: 10.1007/978-1-0716-3690-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
SARS-CoV-2 continues to threaten global public health, making COVID-19 immunity studies of utmost importance. Waning of antibody responses postinfection and/or vaccination and the emergence of immune escape variants have been ongoing challenges in mitigating SARS-CoV-2 morbidity and mortality. While a tremendous amount of work has been done to characterize humoral immune responses to SARS-CoV-2 virus and vaccines, cellular immunity, mediated by T cells, is critical for efficient viral control and protection and demonstrates high durability and cross-reactivity to coronavirus variants. Thus, ELISPOT, a standard assay for antigen-specific cellular immune response assessment, allows us to evaluate SARS-CoV-2-specific T-cell response by quantifying the frequency of SARS-CoV-2-specific cytokine-secreting cells in vitro. We have outlined a detailed procedure to study T-cell recall responses to SARS-CoV-2 in human peripheral blood mononuclear cells (PBMCs) following infection and/or vaccination using an optimized IFN-γ ELISPOT assay. Our methodologies can be adapted to assess other cytokines and are a useful tool for studying other viral pathogen and/or peptide-specific T-cell responses.
Collapse
Affiliation(s)
| | - Ella C Kaufmann
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| | - Tamar Ratishvili
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| | - Ilya M Swanson
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| | | | - Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
44
|
Canevari JT, Cheng AC, Wu L, Rowe SL, Wollersheim DE, West D, Majumdar SS, Sullivan SG. The relative effectiveness of three and four doses of COVID-19 vaccine in Victoria, Australia: A data linkage study. Vaccine 2024; 42:53-58. [PMID: 38057205 DOI: 10.1016/j.vaccine.2023.11.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND The Coronavirus Disease 2019 (COVID-19) pandemic led to extensive vaccination campaigns worldwide, including in Australia. Immunity waning and the emergence of new viral variants pose challenges to the effectiveness of vaccines. Our study aimed to assess the relative effectiveness (rVE) of 3 and 4 compared with 2 doses of COVID-19 vaccine. The study focuses on the Victorian population, a majority of whom had no prior exposure to the virus before vaccination. METHODS We used routinely collected data for the state of Victoria, Australia, to assess rVE during an Omicron-dominant period, 1 June 2022 to 1 March 2023. Immunisation, notifications, hospitalisations and mortality data for residents aged 65 years and older were linked for analysis. Cox proportional hazard regression was used to estimate the rVE against COVID-19 hospitalisation or death, accounting for key confounders with vaccination as a time-varying covariate. RESULTS In 1,070,113 people 65 years or older who had received their second dose, a third and fourth dose of a COVID-19 vaccine significantly reduced the hazard of hospitalisation or death compared to two doses. rVE was highest within two weeks from administration at 40 % (95 % CI: 0 % to 64 %) and 66 % (95 % CI: 60 % to 71 %) for a third and fourth dose, respectively. Additional protection conferred by third and fourth doses waned over time from administration. CONCLUSIONS Our findings underscore the need for additional vaccine doses and updated vaccine strategies. These findings have implications for public health advice and COVID-19 vaccine strategies. Further research and monitoring of vaccine effectiveness in real-world settings are warranted to inform ongoing pandemic response efforts.
Collapse
Affiliation(s)
- Jose T Canevari
- Department of Health, Melbourne, Australia; The University of Melbourne, Melbourne, Australia
| | - Allen C Cheng
- Monash University, Melbourne, Australia; Monash Health, Melbourne Australia
| | - Logan Wu
- Department of Health, Melbourne, Australia; Walter and Eliza Hall Institute, Melbourne, Australia; The University of Melbourne, Melbourne, Australia
| | - Stacey L Rowe
- Department of Health, Melbourne, Australia; Monash University, Melbourne, Australia; University of San Francisco, USA
| | - Dennis E Wollersheim
- Department of Health, Melbourne, Australia; La Trobe University, Melbourne, Australia
| | | | - Suman S Majumdar
- Department of Health, Melbourne, Australia; Monash University, Melbourne, Australia; Burnet Institute, Melbourne, Australia
| | - Sheena G Sullivan
- Department of Health, Melbourne, Australia; WHO Collaborating Centre for Reference and Research on Influenza, Melbourne, Australia; University of California, Los Angeles, USA; The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
45
|
Petersen MS, Pérez-Alós L, í Kongsstovu SK, Eliasen EH, Hansen CB, Larsen S, Hansen JL, Bayarri-Olmos R, Fjallsbak JP, Weihe P, Garred P. Diverging humoral and cellular immune responses due to Omicron-a national study from the Faroe Islands. Microbiol Spectr 2023; 11:e0086523. [PMID: 37909772 PMCID: PMC10714973 DOI: 10.1128/spectrum.00865-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 09/30/2023] [Indexed: 11/03/2023] Open
Abstract
IMPORTANCE The immunity following infection and vaccination with the SARS-CoV-2 Omicron variant is poorly understood. We investigated immunity assessed with antibody and T-cell responses under different scenarios in vaccinated and unvaccinated individuals with and without Omicron infection. We found that the humoral response was higher among vaccinated-naïve than unvaccinated convalescent. Unvaccinated with and without infection had comparable low humoral responses, whereas vaccinated with a second or third dose, independent of infection status, had increasingly higher levels. Only a minor fraction of unvaccinated individuals had detectable humoral responses following Omicron infection, while almost all had positive T-cell responses. In conclusion, primary Omicron infection mounts a low humoral immune response, enhanced by prior vaccination. Omicron infection induced a robust T-cell response in both unvaccinated and vaccinated, demonstrating that immune evasion of primary Omicron infection affects humoral immunity more than T-cell immunity.
Collapse
Affiliation(s)
- Maria Skaalum Petersen
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Eina Hansen Eliasen
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
| | - Cecilie Bo Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Sólrun Larsen
- Chief Medical Officer Office, Tórshavn, Faroe Islands
| | | | - Rafael Bayarri-Olmos
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Recombinant Protein and Antibody Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Pál Weihe
- Department of Research, The National Hospital of the Faroe Islands, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
46
|
Duong S, Burtniak J, Gretchen A, Mai A, Klassen P, Wei Y, Loeppky C, Shaw SY, Bullard J, Van Caeseele P, Stein DR. Riding high: seroprevalence of SARS-CoV-2 after 4 pandemic waves in Manitoba, Canada, April 2020-February 2022. BMC Public Health 2023; 23:2420. [PMID: 38053033 PMCID: PMC10696886 DOI: 10.1186/s12889-023-17239-6] [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: 10/14/2022] [Accepted: 11/16/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND Canada is emerging from the largest SARS-CoV-2 Omicron wave to date, with over 3.3 million confirmed cases. Unfortunately, PCR confirmed cases illuminate only a small portion of infections in the community and underestimate true disease burden. Population based seroprevalence studies, which measure antibody levels against a virus can more accurately estimate infection rates in the community and identify geographical and epidemiological trends to inform public health responses. METHODS The Manitoba COVID-19 Seroprevalence (MCS) study is a population-based cross-sectional study to assess the prevalence of SARS-CoV-2 antibodies across the province. Residual convenience specimens (n = 14,901) were tested for anti-SARS-CoV-2 nucleocapsid and spike IgG antibodies from April 1, 2020 to February 31, 2022. We estimated the monthly and cumulative prevalence using an exponential decay model, accounting for population demographics, sensitivity/specificity, and antibody waning. This approach generated estimates of natural infection as well as total antibody including vaccine-induced immunity within the community. FINDINGS After four waves of the pandemic, 60.1% (95%CI-56.6-63.7) of Manitobans have generated SARS-CoV-2 antibodies due to natural exposure independent of vaccination. Geographical analysis indicates a large portion of provincial prevalence stems from increased transmission in the Northern (92.3%) and Southern (71.8%) regional health authorities. Despite the high mortality rates reported by Manitoba, infection fatality ratios (IFR) peaked at 0.67% and declined to 0.20% following the Omicron wave, indicating parity with other national and international jurisdictions. Manitoba has achieved 93.4% (95%CI- 91.5-95.1) total antibody when including vaccination. INTERPRETATION Our data shows that more than 3 in 5 Manitobans have been infected by SARS-CoV-2 after four waves of the pandemic. This study also identifies key geographical and age specific prevalence rates that have contributed greatly to the overall severity of the pandemic in Manitoba and will inform jurisdictions considering reduction of public health measures.
Collapse
Affiliation(s)
| | | | | | - Anh Mai
- Cadham Provincial Laboratory, Manitoba Health, Winnipeg, Canada
| | - Penny Klassen
- Cadham Provincial Laboratory, Manitoba Health, Winnipeg, Canada
| | - Yichun Wei
- Epidemiology & Surveillance, Manitoba Health, Seniors and Active Living, Winnipeg, Canada
| | - Carla Loeppky
- Epidemiology & Surveillance, Manitoba Health, Seniors and Active Living, Winnipeg, Canada
| | | | - Jared Bullard
- University of Manitoba, Winnipeg, Canada
- Cadham Provincial Laboratory, Manitoba Health, Winnipeg, Canada
| | - Paul Van Caeseele
- University of Manitoba, Winnipeg, Canada
- Cadham Provincial Laboratory, Manitoba Health, Winnipeg, Canada
| | - Derek Riley Stein
- University of Manitoba, Winnipeg, Canada.
- Cadham Provincial Laboratory, Manitoba Health, Winnipeg, Canada.
| |
Collapse
|
47
|
Strizki JM, Grobler JA, Murgolo N, Fridman A, Johnson MG, Du J, Carmelitano P, Brown ML, Paschke A, De Anda C. Virologic Outcomes with Molnupiravir in Non-hospitalized Adult Patients with COVID-19 from the Randomized, Placebo-Controlled MOVe-OUT Trial. Infect Dis Ther 2023; 12:2725-2743. [PMID: 37995070 PMCID: PMC10746688 DOI: 10.1007/s40121-023-00891-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/31/2023] [Indexed: 11/24/2023] Open
Abstract
INTRODUCTION The randomized, placebo-controlled, double-blind MOVe-OUT trial demonstrated molnupiravir (800 mg every 12 h for 5 days) as safe and effective for outpatient treatment of mild-to-moderate COVID-19, significantly reducing the risk of hospitalization/death in high-risk adults. At the time of that report, virologic assessments from the trial were partially incomplete as a result of their time-intensive nature. Here we present final results from all prespecified virology endpoints in MOVe-OUT based on the full trial dataset. METHODS Nasopharyngeal swabs were collected at baseline (day 1, prior to first dose) and days 3, 5 (end-of-treatment visit), 10, 15, and 29. From these samples, change from baseline in SARS-CoV-2 RNA titers (determined by quantitative PCR), detection of infectious SARS-CoV-2 (by plaque assay), and SARS-CoV-2 viral error induction (determined by whole genome next-generation sequencing) were assessed as exploratory endpoints. RESULTS Molnupiravir was associated with greater mean reductions from baseline in SARS-CoV-2 RNA than placebo (including 50% relative reduction at end-of-treatment) through day 10. Among participants with infectious virus detected at baseline (n = 96 molnupiravir, n = 97 placebo) and evaluable post-baseline samples, no molnupiravir-treated participant had infectious SARS-CoV-2 by day 3, whereas infectious virus was recovered from 21% of placebo-arm participants on day 3 and 2% at end-of-treatment. Consistent with molnupiravir's mechanism of action, sequence analysis demonstrated that molnupiravir was associated with an increased number of low-frequency transition errors randomly distributed across the SARS-CoV-2 RNA genome compared with placebo (median 143.5 molnupiravir, 15 placebo), while transversion errors were infrequent overall (median 2 in both arms). Outcomes were consistent regardless of baseline SARS-CoV-2 clade, presence of SARS-CoV-2-specific immune response, or viral load. CONCLUSIONS A 5-day course of orally administered molnupiravir demonstrated a consistently greater virologic effect than placebo, including rapidly eliminating infectious SARS-CoV-2, in high-risk outpatients with mild-to-moderate COVID-19. TRIAL REGISTRATION ClinicalTrials.gov, NCT04575597.
Collapse
Affiliation(s)
| | - Jay A Grobler
- Merck & Co., Inc., 90 E Scott Ave, Rahway, NJ, 07065, USA
| | | | - Arthur Fridman
- Merck & Co., Inc., 90 E Scott Ave, Rahway, NJ, 07065, USA
| | | | - Jiejun Du
- Merck & Co., Inc., 90 E Scott Ave, Rahway, NJ, 07065, USA
| | | | | | - Amanda Paschke
- Merck & Co., Inc., 90 E Scott Ave, Rahway, NJ, 07065, USA
| | - Carisa De Anda
- Merck & Co., Inc., 90 E Scott Ave, Rahway, NJ, 07065, USA
| |
Collapse
|
48
|
Koletzko S, Le Thi TG, Zhelyazkova A, Osterman A, Wichert SP, Breiteneicher S, Koletzko L, Schwerd T, Völk S, Jebrini T, Horak J, Tuschen M, Choukér A, Hornung V, Keppler OT, Koletzko B, Török HP, Adorjan K. A prospective longitudinal cohort study on risk factors for COVID-19 vaccination failure (RisCoin): methods, procedures and characterization of the cohort. Clin Exp Med 2023; 23:4901-4917. [PMID: 37659994 PMCID: PMC10725370 DOI: 10.1007/s10238-023-01170-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/11/2023] [Indexed: 09/04/2023]
Abstract
The primary objective of the RisCoin study was to investigate the interplay of genetic, metabolic, and lifestyle factors as well as stress levels on influencing the humoral immune response after at least two COVID-19 vaccinations, primarily with mRNAs, and the risk of SARS-CoV-2 breakthrough infections during follow-up. Here, we describe the study design, procedures, and study population. RisCoin is a prospective, monocentric, longitudinal, observational cohort study. Between October and December 2021, 4515 participants with at least two COVID-19 vaccinations, primarily BNT162b2 and mRNA-1273, were enrolled at the LMU University Hospital of Munich, thereof > 4000 healthcare workers (HCW), 180 patients with inflammatory bowel disease under immunosuppression, and 119 patients with mental disorders. At enrollment, blood and saliva samples were collected to measure anti-SARS-CoV-2 antibodies, their neutralizing capacity against Omicron-BA.1, stress markers, metabolomics, and genetics. To ensure the confidential handling of sensitive data of study participants, we developed a data protection concept and a mobile application for two-way communication. The application allowed continuous data reporting, including breakthrough infections by the participants, despite irreversible anonymization. Up to 1500 participants attended follow-up visits every two to six months after enrollment. The study gathered comprehensive data and bio-samples of a large representative HCW cohort and two patient groups allowing analyses of complex interactions. Our data protection concept combined with the mobile application proves the feasibility of longitudinal assessment of anonymized participants. Our concept may serve as a blueprint for other studies handling sensitive data on HCW.
Collapse
Affiliation(s)
- Sibylle Koletzko
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstraße 4, 80337, Munich, Germany.
- Department of Pediatrics, Gastroenterology and Nutrition, School of Medicine Collegium, Medicum University of Warmia and Mazury, Olsztyn, Poland.
| | - Thu Giang Le Thi
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstraße 4, 80337, Munich, Germany
| | - Ana Zhelyazkova
- Institut für Notfallmedizin und Medizinmanagement (INM), Klinikum der Universität München, LMU München, Munich, Germany
| | - Andreas Osterman
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site, Munich, Germany
| | - Sven P Wichert
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany
| | | | - Leandra Koletzko
- Department of Medicine II, LMU University Hospital, LMU Munich, Munich, Germany
| | - Tobias Schwerd
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstraße 4, 80337, Munich, Germany
| | - Stefanie Völk
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Tarek Jebrini
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany
| | - Jeannie Horak
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstraße 4, 80337, Munich, Germany
| | - Marina Tuschen
- Department of Anesthesiology, Laboratory of Translational Research Stress and Immunity, LMU University Hospital, LMU Munich, Munich, Germany
| | - Alexander Choukér
- Department of Anesthesiology, Laboratory of Translational Research Stress and Immunity, LMU University Hospital, LMU Munich, Munich, Germany
| | - Veit Hornung
- Gene Center and Department of Biochemistry, LMU Munich, Munich, Germany
| | - Oliver T Keppler
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site, Munich, Germany
| | - Berthold Koletzko
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstraße 4, 80337, Munich, Germany
| | - Helga P Török
- Department of Medicine II, LMU University Hospital, LMU Munich, Munich, Germany
| | - Kristina Adorjan
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany.
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany.
- Center for International Health (CIH), LMU Munich, Munich, Germany.
| |
Collapse
|
49
|
Takizawa T, Ihara K, Uno S, Ohtani S, Watanabe N, Imai N, Nakahara J, Hori S, Garcia-Azorin D, Martelletti P. Metabolic and toxicological considerations regarding CGRP mAbs and CGRP antagonists to treat migraine in COVID-19 patients: a narrative review. Expert Opin Drug Metab Toxicol 2023; 19:951-967. [PMID: 37925645 DOI: 10.1080/17425255.2023.2280221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 11/02/2023] [Indexed: 11/07/2023]
Abstract
INTRODUCTION Migraine pharmacological therapies targeting calcitonin gene-related peptide (CGRP), including monoclonal antibodies and gepants, have shown clinical effect and optimal tolerability. Interactions between treatments of COVID-19 and CGRP-related drugs have not been reviewed. AREAS COVERED An overview of CGRP, a description of the characteristics of each CGRP-related drug and its response predictors, COVID-19 and its treatment, the interactions between CGRP-related drugs and COVID-19 treatment, COVID-19 and vaccination-induced headache, and the neurological consequences of Covid-19. EXPERT OPINION Clinicians should be careful about using gepants for COVID-19 patients, due to the potential drug interactions with drugs metabolized via CYP3A4 cytochrome. In particular, COVID-19 treatment (especially nirmatrelvir packaged with ritonavir, as Paxlovid) should be considered cautiously. It is advisable to stop or adjust the dose (10 mg atogepant when used for episodic migraine) of gepants when using Paxlovid (except for zavegepant). CGRP moncolconal antibodies (CGRP-mAbs) do not have drug - drug interactions, but a few days' interval between a COVID-19 vaccination and the use of CGRP mAbs is recommended to allow the accurate identification of the possible adverse effects, such as injection site reaction. Covid-19- and vaccination-related headache are known to occur. Whether CGRP-related drugs would be of benefit in these circumstances is not yet known.
Collapse
Affiliation(s)
- Tsubasa Takizawa
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Keiko Ihara
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
- Japanese Red Cross Ashikaga Hospital, Ashikaga, Japan
| | - Shunsuke Uno
- Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan
| | - Seiya Ohtani
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
- Division of Drug Informatics, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Narumi Watanabe
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Noboru Imai
- Department of Neurology, Japanese Red Cross Shizuoka Hospital, Shizuoka, Japan
| | - Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Satoko Hori
- Division of Drug Informatics, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - David Garcia-Azorin
- Headache Unit, Department of Neurology, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Paolo Martelletti
- School of Health Sciences, Unitelma Sapienza University of Rome, Rome, Italy
| |
Collapse
|
50
|
Xu Z, Song J, Liu W, Wei D. An agent-based model with antibody dynamics information in COVID-19 epidemic simulation. Infect Dis Model 2023; 8:1151-1168. [PMID: 38033394 PMCID: PMC10685381 DOI: 10.1016/j.idm.2023.11.001] [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: 05/11/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 12/02/2023] Open
Abstract
Accurate prediction of the temporal and spatial characteristics of COVID-19 infection is of paramount importance for effective epidemic prevention and control. In order to accomplish this objective, we incorporated individual antibody dynamics into an agent-based model and devised a methodology that encompasses the dynamic behaviors of each individual, thereby explicitly capturing the count and spatial distribution of infected individuals with varying symptoms at distinct time points. Our model also permits the evaluation of diverse prevention and control measures. Based on our findings, the widespread employment of nucleic acid testing and the implementation of quarantine measures for positive cases and their close contacts in China have yielded remarkable outcomes in curtailing a less transmissible yet more virulent strain; however, they may prove inadequate against highly transmissible and less virulent variants. Additionally, our model excels in its ability to trace back to the initial infected case (patient zero) through early epidemic patterns. Ultimately, our model extends the frontiers of traditional epidemiological simulation methodologies and offers an alternative approach to epidemic modeling.
Collapse
Affiliation(s)
- Zhaobin Xu
- Department of Life Science, Dezhou University, Shandong, 253023, China
| | - Jian Song
- Department of Life Science, Dezhou University, Shandong, 253023, China
| | - Weidong Liu
- Department of Physical Education, Dezhou University, Shandong, 253023, China
| | - Dongqing Wei
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200030, China
| |
Collapse
|