1
|
Houser CL, Fenner KN, Lawrence BP. Timing influences the impact of aryl hydrocarbon receptor activation on the humoral immune response to respiratory viral infection. Toxicol Appl Pharmacol 2024; 489:117010. [PMID: 38901696 PMCID: PMC11240840 DOI: 10.1016/j.taap.2024.117010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024]
Abstract
Humoral responses to respiratory viruses, such as influenza viruses, develop over time and are central to protection from repeated infection with the same or similar viruses. Epidemiological and experimental studies have linked exposures to environmental contaminants that bind the aryl hydrocarbon receptor (AHR) with modulated antibody responses to pathogenic microorganisms and common vaccinations. Other studies have prompted investigation into the potential therapeutic applications of compounds that activate AHR. Herein, using two different AHR ligands [2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and 2-(1H-Indol-3-ylcarbonyl)-4-thiazolecarboxylic acid methyl ester (ITE), to modulate the duration of AHR activity, we show that the humoral response to viral infection is dependent upon the duration and timing of AHR signaling, and that different cellular elements of the response have different sensitivities. When AHR activation was initiated prior to infection with influenza A virus, there was suppression of all measured elements of the humoral response (i.e., the frequency of T follicular helper cells, germinal center B cells, plasma cells, and circulating virus-specific antibody). However, when the timing of AHR activation was adjusted to either early (days -1 to +5 relative to infection) or later (days +5 onwards), then AHR activation affected different aspects of the overall humoral response. These findings highlight the importance of considering the timing of AHR activation in relation to triggering an immune response, particularly when targeting the AHR to manipulate disease processes.
Collapse
Affiliation(s)
- Cassandra L Houser
- Department of Microbiology & Immunology, University of Rochester, Rochester NY14642, USA
| | - Kristina N Fenner
- Department of Environmental Medicine, University of Rochester, Rochester NY14642, USA
| | - B Paige Lawrence
- Department of Microbiology & Immunology, University of Rochester, Rochester NY14642, USA; Department of Environmental Medicine, University of Rochester, Rochester NY14642, USA.
| |
Collapse
|
2
|
Baran J, Kuryk Ł, Szczepińska T, Łaźniewski M, Garofalo M, Mazurkiewicz-Pisarek A, Mikiewicz D, Mazurkiewicz A, Trzaskowski M, Wieczorek M, Pancer K, Hallmann E, Brydak L, Plewczynski D, Ciach T, Mierzejewska J, Staniszewska M. In vitro immune evaluation of adenoviral vector-based platform for infectious diseases. BIOTECHNOLOGIA 2023; 104:403-419. [PMID: 38213479 PMCID: PMC10777723 DOI: 10.5114/bta.2023.132775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 09/12/2023] [Accepted: 09/29/2023] [Indexed: 01/13/2024] Open
Abstract
New prophylactic vaccine platforms are imperative to combat respiratory infections. The efficacy of T and B memory cell-mediated protection, generated through the adenoviral vector, was tested to assess the effectiveness of the new adenoviral-based platforms for infectious diseases. A combination of adenovirus AdV1 (adjuvant), armed with costimulatory ligands (ICOSL and CD40L), and rRBD (antigen: recombinant nonglycosylated spike protein rRBD) was used to promote the differentiation of T and B lymphocytes. Adenovirus AdV2 (adjuvant), without ligands, in combination with rRBD, served as a control. In vitro T-cell responses to the AdV1+rRBD combination revealed that CD8+ platform-specific T-cells increased (37.2 ± 0.7% vs. 23.1 ± 2.1%), and T-cells acted against SARS-CoV-2 via CD8+TEMRA (50.0 ± 1.3% vs. 36.0 ± 3.2%). Memory B cells were induced after treatment with either AdV1+rRBD (84.1 ± 0.8% vs. 82.3 ± 0.4%) or rRBD (94.6 ± 0.3% vs. 82.3 ± 0.4%). Class-switching from IgM and IgD to isotype IgG following induction with rRBD+Ab was observed. RNA-seq profiling identified gene expression patterns related to T helper cell differentiation that protect against pathogens. The analysis determined signaling pathways controlling the induction of protective immunity, including the MAPK cascade, adipocytokine, cAMP, TNF, and Toll-like receptor signaling pathway. The AdV1+rRBD formulation induced IL-6, IL-8, and TNF. RNA-seq of the VERO E6 cell line showed differences in the apoptosis gene expression stimulated with the platforms vs. mock. In conclusion, AdV1+rRBD effectively generates T and B memory cell-mediated protection, presenting promising results in producing CD8+ platform-specific T cells and isotype-switched IgG memory B cells. The platform induces protective immunity by controlling the Th1, Th2, and Th17 cell differentiation gene expression patterns. Further studies are required to confirm its effectiveness.
Collapse
Affiliation(s)
- Joanna Baran
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Łukasz Kuryk
- National Institute of Public Health, Warsaw, Poland
| | - Teresa Szczepińska
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Michał Łaźniewski
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | | | | | - Diana Mikiewicz
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Alina Mazurkiewicz
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Maciej Trzaskowski
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | | | | | | | - Lidia Brydak
- National Institute of Public Health, Warsaw, Poland
| | - Dariusz Plewczynski
- Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
- Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Tomasz Ciach
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | | | - Monika Staniszewska
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| |
Collapse
|
3
|
Thwaites RS, Uruchurtu ASS, Negri VA, Cole ME, Singh N, Poshai N, Jackson D, Hoschler K, Baker T, Scott IC, Ros XR, Cohen ES, Zambon M, Pollock KM, Hansel TT, Openshaw PJM. Early mucosal events promote distinct mucosal and systemic antibody responses to live attenuated influenza vaccine. Nat Commun 2023; 14:8053. [PMID: 38052824 PMCID: PMC10697962 DOI: 10.1038/s41467-023-43842-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023] Open
Abstract
Compared to intramuscular vaccines, nasally administered vaccines have the advantage of inducing local mucosal immune responses that may block infection and interrupt transmission of respiratory pathogens. Live attenuated influenza vaccine (LAIV) is effective in preventing influenza in children, but a correlate of protection for LAIV remains unclear. Studying young adult volunteers, we observe that LAIV induces distinct, compartmentalized, antibody responses in the mucosa and blood. Seeking immunologic correlates of these distinct antibody responses we find associations with mucosal IL-33 release in the first 8 hours post-inoculation and divergent CD8+ and circulating T follicular helper (cTfh) T cell responses 7 days post-inoculation. Mucosal antibodies are induced separately from blood antibodies, are associated with distinct immune responses early post-inoculation, and may provide a correlate of protection for mucosal vaccination. This study was registered as NCT04110366 and reports primary (mucosal antibody) and secondary (blood antibody, and nasal viral load and cytokine) endpoint data.
Collapse
Affiliation(s)
- Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK.
| | | | - Victor Augusti Negri
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Megan E Cole
- Department of Infectious Disease, Imperial College London, London, UK
| | - Nehmat Singh
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Nelisa Poshai
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | - Tina Baker
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ian C Scott
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Xavier Romero Ros
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Emma Suzanne Cohen
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Maria Zambon
- United Kingdom Health Security Agency, London, UK
| | - Katrina M Pollock
- Department of Infectious Disease, Imperial College London, London, UK
| | - Trevor T Hansel
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
4
|
Moreno-Millán N, Botanes Iglesias J, Robles Raya P. [Vaccine strategy in hibernal viruses: Present and future]. Aten Primaria 2023; 55:102674. [PMID: 37262974 PMCID: PMC10242488 DOI: 10.1016/j.aprim.2023.102674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023] Open
Affiliation(s)
- Nemesio Moreno-Millán
- Sistemes d'informació dels Serveis d'Atenció Primària (SISAP), Institut Català de la Salut (ICS), Barcelona, España; Miembro del Grupo de Vacunas y Profilaxis de enfermedades infecciosas de la Societat Catalana de Medicina de Família i Comunitària (CAMFiC)
| | - Jenifer Botanes Iglesias
- CAP Can Moritz, Institut Català de la Salut (ICS), Cornellà de Llobregat, Barcelona, España; Miembro del Grupo de Vacunas y Profilaxis de enfermedades infecciosas de la Societat Catalana de Medicina de Família i Comunitària (CAMFiC).
| | - Purificación Robles Raya
- CAP Can Vidalet, Institut Català de la Salut (ICS), Esplugues de LLobregat, Barcelona, España; Miembro del Grupo de Vacunas y Profilaxis de enfermedades infecciosas de la Societat Catalana de Medicina de Família i Comunitària (CAMFiC)
| |
Collapse
|
5
|
Moore KA, Leighton T, Ostrowsky JT, Anderson CJ, Danila RN, Ulrich AK, Lackritz EM, Mehr AJ, Baric RS, Baylor NW, Gellin BG, Gordon JL, Krammer F, Perlman S, Rees HV, Saville M, Weller CL, Osterholm MT. A research and development (R&D) roadmap for broadly protective coronavirus vaccines: A pandemic preparedness strategy. Vaccine 2023; 41:2101-2112. [PMID: 36870874 PMCID: PMC9941884 DOI: 10.1016/j.vaccine.2023.02.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023]
Abstract
Broadly protective coronavirus vaccines are an important tool for protecting against future SARS-CoV-2 variants and could play a critical role in mitigating the impact of future outbreaks or pandemics caused by novel coronaviruses. The Coronavirus Vaccines Research and Development (R&D) Roadmap (CVR) is aimed at promoting the development of such vaccines. The CVR, funded by the Bill & Melinda Gates Foundation and The Rockefeller Foundation, was generated through a collaborative and iterative process, which was led by the Center for Infectious Disease Research and Policy (CIDRAP) at the University of Minnesota and involved 50 international subject matter experts and recognized leaders in the field. This report summarizes the major issues and areas of research outlined in the CVR and identifies high-priority milestones. The CVR covers a 6-year timeframe and is organized into five topic areas: virology, immunology, vaccinology, animal and human infection models, and policy and finance. Included in each topic area are key barriers, gaps, strategic goals, milestones, and additional R&D priorities. The roadmap includes 20 goals and 86 R&D milestones, 26 of which are ranked as high priority. By identifying key issues, and milestones for addressing them, the CVR provides a framework to guide funding and research campaigns that promote the development of broadly protective coronavirus vaccines.
Collapse
Affiliation(s)
- Kristine A Moore
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA; Center for Infectious Disease Research and Policy, C315 Mayo Memorial Building, MMC 263, 420 Delaware Street, SE, Minneapolis, Minnesota 55455, USA.
| | - Tabitha Leighton
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Julia T Ostrowsky
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cory J Anderson
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Angela K Ulrich
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Eve M Lackritz
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Angela J Mehr
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ralph S Baric
- University of North Carolina, Chapel Hill, North Carolina, USA
| | | | | | - Jennifer L Gordon
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Florian Krammer
- Department of Microbiology, Department of Pathology, Molecular and Cell-Based Medicine, and Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | | | - Helen V Rees
- Wits RHI, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Melanie Saville
- Coalition for Epidemic Preparedness Innovations, London, United Kingdom
| | | | - Michael T Osterholm
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
6
|
Parker E, Thomas J, Roper KJ, Ijaz S, Edwards T, Marchesin F, Katsanovskaja K, Lett L, Jones C, Hardwick HE, Davis C, Vink E, McDonald SE, Moore SC, Dicks S, Jegatheesan K, Cook NJ, Hope J, Cherepanov P, McClure MO, Baillie JK, Openshaw PJM, Turtle L, Ho A, Semple MG, Paxton WA, Tedder RS, Pollakis G. SARS-CoV-2 antibody responses associate with sex, age and disease severity in previously uninfected people admitted to hospital with COVID-19: An ISARIC4C prospective study. Front Immunol 2023; 14:1146702. [PMID: 37056776 PMCID: PMC10087108 DOI: 10.3389/fimmu.2023.1146702] [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: 01/17/2023] [Accepted: 02/27/2023] [Indexed: 03/17/2023] Open
Abstract
The SARS-CoV-2 pandemic enables the analysis of immune responses induced against a novel coronavirus infecting immunologically naïve individuals. This provides an opportunity for analysis of immune responses and associations with age, sex and disease severity. Here we measured an array of solid-phase binding antibody and viral neutralising Ab (nAb) responses in participants (n=337) of the ISARIC4C cohort and characterised their correlation with peak disease severity during acute infection and early convalescence. Overall, the responses in a Double Antigen Binding Assay (DABA) for antibody to the receptor binding domain (anti-RBD) correlated well with IgM as well as IgG responses against viral spike, S1 and nucleocapsid protein (NP) antigens. DABA reactivity also correlated with nAb. As we and others reported previously, there is greater risk of severe disease and death in older men, whilst the sex ratio was found to be equal within each severity grouping in younger people. In older males with severe disease (mean age 68 years), peak antibody levels were found to be delayed by one to two weeks compared with women, and nAb responses were delayed further. Additionally, we demonstrated that solid-phase binding antibody responses reached higher levels in males as measured via DABA and IgM binding against Spike, NP and S1 antigens. In contrast, this was not observed for nAb responses. When measuring SARS-CoV-2 RNA transcripts (as a surrogate for viral shedding) in nasal swabs at recruitment, we saw no significant differences by sex or disease severity status. However, we have shown higher antibody levels associated with low nasal viral RNA indicating a role of antibody responses in controlling viral replication and shedding in the upper airway. In this study, we have shown discernible differences in the humoral immune responses between males and females and these differences associate with age as well as with resultant disease severity.
Collapse
Affiliation(s)
- Eleanor Parker
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Jordan Thomas
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Kelly J. Roper
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Samreen Ijaz
- Blood Borne Virus Unit, Reference Department, UK Health Security Agency, London, United Kingdom
| | - Tansy Edwards
- Medical Research Council (MRC) International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Federica Marchesin
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Ksenia Katsanovskaja
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Lauren Lett
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Christopher Jones
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Hayley E. Hardwick
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Chris Davis
- Medical Research Council, University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Elen Vink
- Medical Research Council, University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Sarah E. McDonald
- Medical Research Council, University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Shona C. Moore
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Steve Dicks
- Blood Borne Virus Unit, Reference Department, UK Health Security Agency, London, United Kingdom
- National Health Service (NHS) Blood and Transplant, London, United Kingdom
| | - Keerthana Jegatheesan
- Blood Borne Virus Unit, Reference Department, UK Health Security Agency, London, United Kingdom
- National Health Service (NHS) Blood and Transplant, London, United Kingdom
| | - Nicola J. Cook
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Joshua Hope
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Peter Cherepanov
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Myra O. McClure
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | | | | | - Lance Turtle
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Antonia Ho
- Medical Research Council, University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Malcolm G. Semple
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - William A. Paxton
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Richard S. Tedder
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Georgios Pollakis
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | | |
Collapse
|
7
|
Ivanova I, Filippenko A, Trufanova A, Omelchenko N, Chemisova O, Vodopyanov A, Bereznyak E, Sokolova E, Noskov A, Totolyan AAA. Assessment of the formation and intensity of adaptive immunity in patients with COVID-19. RUSSIAN JOURNAL OF INFECTION AND IMMUNITY 2023. [DOI: 10.15789/2220-7619-aof-2107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
The study of adaptive immunity in survivors of a new coronavirus infection is an important task, since there is no consensus on whether the severity of the disease affects the formation and intensity of the immune response to COVID-19. In this regard, a comparative assessment of the presence and duration of preservation of cellular and humoral immunity in patients with COVID-19 of varying severity was carried out. The study involved volunteers who had been ill with a new coronavirus infection asymptomatically (n=30), in moderate severity (n=21) and in severe form (n=12). The average age of the subjects was 47.312.5 years. The formation of cellular immunity was judged by an increase in the synthesis of IFN- in response to stimulation of lymphocytes for 16-20 hours by glycoprotein S(RBD) of the causative agent COVID-19. To determine IFN- products, the gamma InterferonIFA-BEST test system, manufactured by Vector-Best JSC, Russia, was used. The humoral immune response was recorded by detecting class G antibodies using the "SARS-CoV-2RBD-ELISA-Gamalei" test system (FSBI "N.F. Gamalei NITSEM" of the Ministry of Health of the Russian Federation). The results obtained indicate that patients from all groups have both humoral and cellular immunity to the causative agent of a new coronavirus infection. However, the number of people with adaptive immunity to COVID-19 and the duration of its preservation depends on the severity of the infection. A significant decrease in the number of people with cellular immunity was revealed in the group of seriously ill. At the same time, the majority of volunteers in this group registered the presence of class G immunoglobulins before the end of observation. In this group, unlike the other two, no patients were identified in whom only the cellular link of the immune response was activated. Volunteers who did not retain adaptive immunity to the causative agent of a new coronavirus infection appeared only by the end of the observation period. Among those who had the disease in an average form 7-8 months after recovery, there was a decrease in the number of people with cellular and humoral immunity. This process started earlier than in the group of patients who were asymptomatic and continued until the end of the study. The proportion of individuals with cellular immunity increased, and at a later date with a humoral immune response. By the end of the study, a high percentage of volunteers remained asymptomatically infected, having cellular and humoral immunity to SARS-CoV-2. Their number remained statistically higher than in the group of those who had a new coronavirus infection of moderate severity, but lower than in the group of those who were seriously ill. Also in this group, by the end of the experiment, an increase in the number of volunteers with only a cellular immune response was recorded. According to the data obtained, at the end of the observation period, the number of volunteers with humoral immunity to the causative agent of a new coronavirus infection is higher compared to those with a cellular immune response
Collapse
|
8
|
Impact of Reinfection with SARS-CoV-2 Omicron Variants in Previously Infected Hamsters. J Virol 2023; 97:e0136622. [PMID: 36633406 PMCID: PMC9888231 DOI: 10.1128/jvi.01366-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The diversity of SARS-CoV-2 mutations raises the possibility of reinfection of individuals previously infected with earlier variants, and this risk is further increased by the emergence of the B.1.1.529 Omicron variant. In this study, we used an in vivo, hamster infection model to assess the potential for individuals previously infected with SARS-CoV-2 to be reinfected with Omicron variant and we also investigated the pathology associated with such infections. Initially, Syrian hamsters were inoculated with a lineage A, B.1.1.7, B.1.351, B.1.617.2 or a subvariant of Omicron, BA.1 strain and then reinfected with the BA.1 strain 5 weeks later. Subsequently, the impact of reinfection with Omicron subvariants (BA.1 and BA.2) in individuals previously infected with the BA.1 strain was examined. Although viral infection and replication were suppressed in both the upper and lower airways, following reinfection, virus-associated RNA was detected in the airways of most hamsters. Viral replication was more strongly suppressed in the lower respiratory tract than in the upper respiratory tract. Consistent amino acid substitutions were observed in the upper respiratory tract of infected hamsters after primary infection with variant BA.1, whereas diverse mutations appeared in hamsters reinfected with the same variant. Histopathology showed no acute pneumonia or disease enhancement in any of the reinfection groups and, in addition, the expression of inflammatory cytokines and chemokines in the airways of reinfected animals was only mildly elevated. These findings are important for understanding the risk of reinfection with new variants of SARS-CoV-2. IMPORTANCE The emergence of SARS-CoV-2 variants and the widespread use of COVID-19 vaccines has resulted in individual differences in immune status against SARS-CoV-2. A decay in immunity over time and the emergence of variants that partially evade the immune response can also lead to reinfection. In this study, we demonstrated that, in hamsters, immunity acquired following primary infection with previous SARS-CoV-2 variants was effective in preventing the onset of pneumonia after reinfection with the Omicron variant. However, viral infection and multiplication in the upper respiratory tract were still observed after reinfection. We also showed that more diverse nonsynonymous mutations appeared in the upper respiratory tract of reinfected hamsters that had acquired immunity from primary infection. This hamster model reveals the within-host evolution of SARS-CoV-2 and its pathology after reinfection, and provides important information for countermeasures against diversifying SARS-CoV-2 variants.
Collapse
|
9
|
Aksyuk AA, Bansal H, Wilkins D, Stanley AM, Sproule S, Maaske J, Sanikommui S, Hartman WR, Sobieszczyk ME, Falsey AR, Kelly EJ. AZD1222-induced nasal antibody responses are shaped by prior SARS-CoV-2 infection and correlate with virologic outcomes in breakthrough infection. Cell Rep Med 2023; 4:100882. [PMID: 36610390 PMCID: PMC9750884 DOI: 10.1016/j.xcrm.2022.100882] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/11/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
The nasal mucosa is an important initial site of host defense against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. However, intramuscularly administered vaccines typically do not achieve high antibody titers in the nasal mucosa. We measure anti-SARS-CoV-2 spike immunoglobulin G (IgG) and IgA in nasal epithelial lining fluid (NELF) following intramuscular vaccination of 3,058 participants from the immunogenicity substudy of a phase 3, double-blind, placebo-controlled study of AZD1222 vaccination (ClinicalTrials.gov: NCT04516746). IgG is detected in NELF collected 14 days following the first AZD1222 vaccination. IgG levels increase with a second vaccination and exceed pre-existing levels in baseline-SARS-CoV-2-seropositive participants. Nasal IgG responses are durable and display strong correlations with serum IgG, suggesting serum-to-NELF transudation. AZD1222 induces short-lived increases to pre-existing nasal IgA levels in baseline-seropositive vaccinees. Vaccinees display a robust recall IgG response upon breakthrough infection, with overall magnitudes unaffected by time between vaccination and illness. Mucosal responses correlate with reduced viral loads and shorter durations of viral shedding in saliva.
Collapse
Affiliation(s)
- Anastasia A Aksyuk
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Himanshu Bansal
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Deidre Wilkins
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Ann Marie Stanley
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Stephanie Sproule
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Jill Maaske
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Satya Sanikommui
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - William R Hartman
- Department of Anesthesiology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53726, USA
| | - Magdalena E Sobieszczyk
- Division of Infectious Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, New York Presbyterian/Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ann R Falsey
- University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Rochester Regional Health, Rochester, NY 14621, USA.
| | - Elizabeth J Kelly
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA.
| |
Collapse
|
10
|
Adaptive Immunity to Viruses: What Did We Learn from SARS-CoV-2 Infection? Int J Mol Sci 2022; 23:ijms232213951. [PMID: 36430430 PMCID: PMC9694482 DOI: 10.3390/ijms232213951] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
The SARS-CoV-2 virus causes various conditions, from asymptomatic infection to the fatal coronavirus disease 2019 (COVID-19). An intact immune system can overcome SARS-CoV-2 and other viral infections. Defective natural, mainly interferon I- and III-dependent, responses may lead to the spread of the virus to multiple organs. Adaptive B- and T-cell responses, including memory, highly influence the severity and outcome of COVID-19. With respect to B-cell immunity, germinal centre formation is delayed or even absent in the most severe cases. Extrafollicular low-affinity anti-SARS-CoV-2 antibody production will occur instead of specific, high-affinity antibodies. Helper and CD8+ cytotoxic T-cells become hyperactivated and then exhausted, leading to ineffective viral clearance from the body. The dysregulation of neutrophils and monocytes/macrophages, as well as lymphocyte hyperreactivity, might lead to the robust production of inflammatory mediators, also known as cytokine storm. Eventually, the disruption of this complex network of immune cells and mediators leads to severe, sometimes fatal COVID-19 or another viral disease.
Collapse
|
11
|
Navaratnam AMD, Shrotri M, Nguyen V, Braithwaite I, Beale S, Byrne TE, Fong WLE, Fragaszy E, Geismar C, Hoskins S, Kovar J, Patel P, Yavlinsky A, Aryee A, Rodger A, Hayward AC, Aldridge RW. Nucleocapsid and spike antibody responses following virologically confirmed SARS-CoV-2 infection: an observational analysis in the Virus Watch community cohort. Int J Infect Dis 2022; 123:104-111. [PMID: 35987470 PMCID: PMC9385348 DOI: 10.1016/j.ijid.2022.07.053] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Seroprevalence studies can provide a measure of SARS-CoV-2 cumulative incidence, but a better understanding of spike and nucleocapsid (anti-N) antibody dynamics following infection is needed to assess the longevity of detectability. METHODS Adults aged ≥18 years, from households enrolled in the Virus Watch prospective community cohort study in England and Wales, provided monthly capillary blood samples, which were tested for spike antibody and anti-N. Participants self-reported vaccination dates and past medical history. Previous polymerase chain reaction (PCR) swabs were obtained through Second Generation Surveillance System linkage data. The primary outcome variables were seropositivity and total anti-N and spike antibody levels after PCR-confirmed infection. RESULTS A total of 13,802 eligible individuals provided 58,770 capillary blood samples. A total of 537 of these had a previous positive PCR-confirmed SARS-CoV-2 infection within 0-269 days of antibody sample date, among them 432 (80.45%) having a positive anti-N result. Median anti-N levels peaked between days 90 and 119 after PCR results and then began to decline. There is evidence of anti-N waning from 120 days onwards, with earlier waning for females and younger age categories. CONCLUSION Our findings suggest that anti-N has around 80% sensitivity for identifying previous COVID-19 infection, and the duration of detectability is affected by sex and age.
Collapse
Affiliation(s)
| | - Madhumita Shrotri
- Institute of Health Informatics, University College London, United Kingdom
| | - Vincent Nguyen
- Institute of Health Informatics, University College London, United Kingdom
| | - Isobel Braithwaite
- Institute of Health Informatics, University College London, United Kingdom
| | - Sarah Beale
- Institute of Epidemiology and Health Care, University College London, London, United Kingdom
| | - Thomas E Byrne
- Institute of Health Informatics, University College London, United Kingdom
| | | | - Ellen Fragaszy
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London, United Kingdom
| | - Cyril Geismar
- Institute of Health Informatics, University College London, United Kingdom
| | - Susan Hoskins
- Institute of Health Informatics, University College London, United Kingdom
| | - Jana Kovar
- Institute of Epidemiology and Health Care, University College London, London, United Kingdom
| | - Parth Patel
- Institute of Health Informatics, University College London, United Kingdom
| | - Alexei Yavlinsky
- Institute of Health Informatics, University College London, United Kingdom
| | - Anna Aryee
- Institute of Health Informatics, University College London, United Kingdom
| | - Alison Rodger
- Institute for Global Health, University College London, London, United Kingdom
| | - Andrew C Hayward
- Institute of Epidemiology and Health Care, University College London, London, United Kingdom
| | - Robert W Aldridge
- Institute of Health Informatics, University College London, United Kingdom.
| |
Collapse
|
12
|
Mahla RS, Dustin LB. Lessons from a large-scale COVID-19 vaccine trial. J Clin Invest 2022; 132:163202. [PMID: 36106630 PMCID: PMC9473721 DOI: 10.1172/jci163202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Global vaccination coverage remains indispensable in combatting the ongoing SARS-CoV-2 pandemic. Safety, efficacy, and durability of immune protection are the key parameters of randomized controlled trials (RCTs) and are essential for vaccine approvals, global distribution, and comprehensive population-vaccination programs. Immune protection from either vaccination or natural infection decreases over time, further challenged by rapid viral evolution. In this issue of the JCI, Sobieszczyk and colleagues report an update on the safety, efficacy, and durability of immune protection of AZD12222 in a large-scale, multinational, Phase III RCT. They report that protection lasted through 6 months, with immunity waning after 180 days. The study also highlights challenges facing vaccine trials, including the need for early unblinding for vulnerable participants, which may affect outcome measurements. Another challenge is to ensure fair representation of marginalized and minority ethnic groups in vaccine safety and efficacy studies worldwide.
Collapse
|
13
|
Yu ED, Narowski TM, Wang E, Garrigan E, Mateus J, Frazier A, Weiskopf D, Grifoni A, Premkumar L, da Silva Antunes R, Sette A. Immunological memory to common cold coronaviruses assessed longitudinally over a three-year period pre-COVID19 pandemic. Cell Host Microbe 2022; 30:1269-1278.e4. [PMID: 35932763 PMCID: PMC9296686 DOI: 10.1016/j.chom.2022.07.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/12/2022] [Accepted: 07/15/2022] [Indexed: 12/01/2022]
Abstract
The immune memory to common cold coronaviruses (CCCs) influences SARS-CoV-2 infection outcome, and understanding its effect is crucial for pan-coronavirus vaccine development. We performed a longitudinal analysis of pre-COVID19-pandemic samples from 2016-2019 in young adults and assessed CCC-specific CD4+ T cell and antibody responses. Notably, CCC responses were commonly detected with comparable frequencies as with other common antigens and were sustained over time. CCC-specific CD4+ T cell responses were associated with low HLA-DR+CD38+ signals, and their magnitude did not correlate with yearly CCC infection prevalence. Similarly, CCC-specific and spike RBD-specific IgG responses were stable in time. Finally, high CCC-specific CD4+ T cell reactivity, but not antibody titers, was associated with pre-existing SARS-CoV-2 immunity. These results provide a valuable reference for understanding the immune response to endemic coronaviruses and suggest that steady and sustained CCC responses are likely from a stable pool of memory CD4+ T cells due to repeated earlier exposures and possibly occasional reinfections.
Collapse
Affiliation(s)
- Esther Dawen Yu
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Tara M Narowski
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7290, USA
| | - Eric Wang
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Emily Garrigan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Jose Mateus
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - April Frazier
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7290, USA
| | - Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA.
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| |
Collapse
|
14
|
Keuning MW, Grobben M, Bijlsma MW, Anker B, Berman-de Jong EP, Cohen S, Felderhof M, de Groen AE, de Groof F, Rijpert M, van Eijk HWM, Tejjani K, van Rijswijk J, Steenhuis M, Rispens T, Plötz FB, van Gils MJ, Pajkrt D. Differences in systemic and mucosal SARS-CoV-2 antibody prevalence in a prospective cohort of Dutch children. Front Immunol 2022; 13:976382. [PMID: 36159841 PMCID: PMC9500453 DOI: 10.3389/fimmu.2022.976382] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Background As SARS-CoV-2 will likely continue to circulate, low-impact methods become more relevant to monitor antibody-mediated immunity. Saliva sampling could provide a non-invasive method with reduced impact on children. Studies reporting on the differences between systemic and mucosal humoral immunity to SARS-CoV-2 are inconsistent in adults and scarce in children. These differences may be further unraveled by exploring associations to demographic and clinical variables. Methods To evaluate the use of saliva antibody assays, we performed a cross-sectional cohort study by collecting serum and saliva of 223 children attending medical services in the Netherlands (irrespective of SARS-CoV-2 exposure, symptoms or vaccination) from May to October 2021. With a Luminex and a Wantai assay, we measured prevalence of SARS-CoV-2 spike (S), receptor binding domain (RBD) and nucleocapsid-specific IgG and IgA in serum and saliva and explored associations with demographic variables. Findings The S-specific IgG prevalence was higher in serum 39% (95% CI 32 - 45%) than in saliva 30% (95% CI 24 - 36%) (P ≤ 0.003). Twenty-seven percent (55/205) of children were S-specific IgG positive in serum and saliva, 12% (25/205) were only positive in serum and 3% (6/205) only in saliva. Vaccinated children showed a higher concordance between serum and saliva than infected children. Odds for saliva S-specific IgG positivity were higher in girls compared to boys (aOR 2.63, P = 0.012). Moreover, immunocompromised children showed lower odds for S- and RBD-specific IgG in both serum and saliva compared to healthy children (aOR 0.23 - 0.25, P ≤ 0.050). Conclusions We showed that saliva-based antibody assays can be useful for identifying SARS-CoV-2 humoral immunity in a non-invasive manner, and that IgG prevalence may be affected by sex and immunocompromisation. Differences between infection and vaccination, between sexes and between immunocompromised and healthy children should be further investigated and considered when choosing systemic or mucosal antibody measurement.
Collapse
Affiliation(s)
- Maya W. Keuning
- Department of Pediatric Infectious Diseases, Rheumatology & Immunology, Amsterdam University Medical Centers (UMC), location University of Amsterdam, Amsterdam, Netherlands
| | - Marloes Grobben
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam University Medical Centers (UMC) location University of Amsterdam, Amsterdam, Netherlands
- Infectious diseases, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Merijn W. Bijlsma
- Department of Pediatrics, Emma Children’s Hospital Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Beau Anker
- Department of Pediatrics, Emma Children’s Hospital Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Eveline P. Berman-de Jong
- Department of Pediatrics, Emma Children’s Hospital Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Sophie Cohen
- Department of Pediatrics, Emma Children’s Hospital Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
| | | | - Anne-Elise de Groen
- Department of Pediatric Infectious Diseases, Rheumatology & Immunology, Amsterdam University Medical Centers (UMC), location University of Amsterdam, Amsterdam, Netherlands
| | - Femke de Groof
- Department of Pediatrics, Noordwest Ziekenhuisgroep, Alkmaar, Netherlands
| | - Maarten Rijpert
- Department of Pediatrics, Zaans Medical Center, Zaandam, Netherlands
| | - Hetty W. M. van Eijk
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam University Medical Centers (UMC) location University of Amsterdam, Amsterdam, Netherlands
| | - Khadija Tejjani
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam University Medical Centers (UMC) location University of Amsterdam, Amsterdam, Netherlands
- Infectious diseases, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Jacqueline van Rijswijk
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam University Medical Centers (UMC) location University of Amsterdam, Amsterdam, Netherlands
- Infectious diseases, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Maurice Steenhuis
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Frans B. Plötz
- Department of Pediatrics, Emma Children’s Hospital Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
- Department of Pediatrics, Tergooi Medical Center, Blaricum, Netherlands
| | - Marit J. van Gils
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam University Medical Centers (UMC) location University of Amsterdam, Amsterdam, Netherlands
- Infectious diseases, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Dasja Pajkrt
- Department of Pediatric Infectious Diseases, Rheumatology & Immunology, Amsterdam University Medical Centers (UMC), location University of Amsterdam, Amsterdam, Netherlands
- Infectious diseases, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| |
Collapse
|
15
|
Plotkin S. Serologic Tests for COVID-19 Infections and Vaccination. Pediatr Infect Dis J 2022; 41:e304-e305. [PMID: 35703283 PMCID: PMC9281429 DOI: 10.1097/inf.0000000000003574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/23/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Stanley Plotkin
- From the Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Pennsylvania, USA
| |
Collapse
|
16
|
Epsi NJ, Richard SA, Lindholm DA, Mende K, Ganesan A, Huprikar N, Lalani T, Fries AC, Maves RC, Colombo RE, Larson DT, Smith A, Chi SW, Maldonado CJ, Ewers EC, Jones MU, Berjohn CM, Libraty DH, Edwards MS, English C, Rozman JS, Mody RM, Colombo CJ, Samuels EC, Nwachukwu P, Tso MS, Scher AI, Byrne C, Rusiecki J, Simons MP, Tribble D, Broder CC, Agan BK, Burgess TH, Laing ED, Pollett SD. Understanding "Hybrid Immunity": Comparison and Predictors of Humoral Immune Responses to Severe Acute Respiratory Syndrome Coronavirus 2 Infection (SARS-CoV-2) and Coronavirus Disease 2019 (COVID-19) Vaccines. Clin Infect Dis 2022; 76:e439-e449. [PMID: 35608504 PMCID: PMC9213853 DOI: 10.1093/cid/ciac392] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/29/2022] [Accepted: 05/17/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Comparison of humoral responses in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccinees, those with SARS-CoV-2 infection, or combinations of vaccine/ infection ("hybrid immunity") may clarify predictors of vaccine immunogenicity. METHODS We studied 2660 US Military Health System beneficiaries with a history of SARS-CoV-2 infection-alone (n = 705), vaccination-alone (n = 932), vaccine-after-infection (n = 869), and vaccine-breakthrough-infection (n = 154). Peak anti-spike-immunoglobulin G (IgG) responses through 183 days were compared, with adjustment for vaccine product, demography, and comorbidities. We excluded those with evidence of clinical or subclinical SARS-CoV-2 reinfection from all groups. RESULTS Multivariable regression results indicated that vaccine-after-infection anti-spike-IgG responses were higher than infection-alone (P < .01), regardless of prior infection severity. An increased time between infection and vaccination was associated with greater post-vaccination IgG response (P < .01). Vaccination-alone elicited a greater IgG response but more rapid waning of IgG (P < .01) compared with infection-alone (P < .01). BNT162b2 and mRNA-1273 vaccine-receipt was associated with greater IgG responses compared with JNJ-78436735 vaccine-receipt (P < .01), regardless of infection history. Those with vaccine-after-infection or vaccine-breakthrough-infection had a more durable anti-spike-IgG response compared to infection-alone (P < .01). CONCLUSIONS Vaccine-receipt elicited higher anti-spike-IgG responses than infection-alone, although IgG levels waned faster in those vaccinated (compared to infection-alone). Vaccine-after-infection elicits a greater humoral response compared with vaccine or infection alone; and the timing, but not disease severity, of prior infection predicted these post-vaccination IgG responses. While differences between groups were small in magnitude, these results offer insights into vaccine immunogenicity variations that may help inform vaccination timing strategies.
Collapse
Affiliation(s)
- Nusrat J Epsi
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
| | - Stephanie A Richard
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
| | - David A Lindholm
- Brooke Army Medical Center, Fort Sam Houston, Texas, USA,Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Katrin Mende
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA,Brooke Army Medical Center, Fort Sam Houston, Texas, USA
| | - Anuradha Ganesan
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA,Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Nikhil Huprikar
- Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Tahaniyat Lalani
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA,Naval Medical Center Portsmouth, Portsmouth, Virginia, USA
| | - Anthony C Fries
- US Air Force School of Aerospace Medicine, Dayton, Ohio, USA
| | - Ryan C Maves
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Rhonda E Colombo
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA,Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Madigan Army Medical Center, Joint Base Lewis McChord, Washington, USA
| | - Derek T Larson
- Fort Belvoir Community Hospital, Fort Belvoir, Virginia, USA,Naval Medical Center San Diego, San Diego, California, USA
| | - Alfred Smith
- Naval Medical Center Portsmouth, Portsmouth, Virginia, USA
| | - Sharon W Chi
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA,Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | | | - Evan C Ewers
- Fort Belvoir Community Hospital, Fort Belvoir, Virginia, USA
| | | | - Catherine M Berjohn
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Naval Medical Center San Diego, San Diego, California, USA
| | - Daniel H Libraty
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA,Naval Medical Center San Diego, San Diego, California, USA
| | - Margaret Sanchez Edwards
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
| | - Caroline English
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
| | - Julia S Rozman
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
| | - Rupal M Mody
- William Beaumont Army Medical Center, El Paso, Texas, USA
| | - Christopher J Colombo
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Madigan Army Medical Center, Joint Base Lewis McChord, Washington, USA
| | - Emily C Samuels
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Princess Nwachukwu
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Marana S Tso
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Ann I Scher
- Department of Preventive Medicine & Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Celia Byrne
- Department of Preventive Medicine & Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jennifer Rusiecki
- Department of Preventive Medicine & Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Mark P Simons
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - David Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Christopher C Broder
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Brian K Agan
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
| | - Timothy H Burgess
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Eric D Laing
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Simon D Pollett
- Correspondence: Simon Pollett, MBBS, 6720A Rockledge Drive, Suite 250, Bethesda, MD 20817, USA ()
| | | |
Collapse
|
17
|
Stewart A, Sinclair E, Ng JCF, O’Hare JS, Page A, Serangeli I, Margreitter C, Orsenigo F, Longman K, Frampas C, Costa C, Lewis HM, Kasar N, Wu B, Kipling D, Openshaw PJM, Chiu C, Baillie JK, Scott JT, Semple MG, Bailey MJ, Fraternali F, Dunn-Walters DK. Pandemic, Epidemic, Endemic: B Cell Repertoire Analysis Reveals Unique Anti-Viral Responses to SARS-CoV-2, Ebola and Respiratory Syncytial Virus. Front Immunol 2022; 13:807104. [PMID: 35592326 PMCID: PMC9111746 DOI: 10.3389/fimmu.2022.807104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/15/2022] [Indexed: 11/17/2022] Open
Abstract
Immunoglobulin gene heterogeneity reflects the diversity and focus of the humoral immune response towards different infections, enabling inference of B cell development processes. Detailed compositional and lineage analysis of long read IGH repertoire sequencing, combining examples of pandemic, epidemic and endemic viral infections with control and vaccination samples, demonstrates general responses including increased use of IGHV4-39 in both Zaire Ebolavirus (EBOV) and COVID-19 patient cohorts. We also show unique characteristics absent in Respiratory Syncytial Virus or yellow fever vaccine samples: EBOV survivors show unprecedented high levels of class switching events while COVID-19 repertoires from acute disease appear underdeveloped. Despite the high levels of clonal expansion in COVID-19 IgG1 repertoires there is a striking lack of evidence of germinal centre mutation and selection. Given the differences in COVID-19 morbidity and mortality with age, it is also pertinent that we find significant differences in repertoire characteristics between young and old patients. Our data supports the hypothesis that a primary viral challenge can result in a strong but immature humoral response where failures in selection of the repertoire risk off-target effects.
Collapse
Affiliation(s)
- Alexander Stewart
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Emma Sinclair
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Joseph Chi-Fung Ng
- Randall Centre for Cell & Molecular Biophysics, King’s College London, London, United Kingdom
| | - Joselli Silva O’Hare
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
- Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Audrey Page
- Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Ilaria Serangeli
- Dipartimento di Biologia e Biotecnologie Charles Darwin, Sapienza Università di Roma, Rome, Italy
| | | | - Federica Orsenigo
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
- Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Katherine Longman
- Department of Chemistry, University of Surrey, Guildford, United Kingdom
| | - Cecile Frampas
- Department of Chemistry, University of Surrey, Guildford, United Kingdom
| | - Catia Costa
- Department of Chemistry, University of Surrey, Guildford, United Kingdom
| | - Holly-May Lewis
- Department of Chemistry, University of Surrey, Guildford, United Kingdom
| | - Nora Kasar
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Bryan Wu
- Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - David Kipling
- School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Peter JM Openshaw
- Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Christopher Chiu
- Faculty of Medicine, Imperial College London, London, United Kingdom
| | - J Kenneth Baillie
- Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Janet T. Scott
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, United Kingdom
| | - Malcolm G. Semple
- Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Melanie J. Bailey
- Department of Chemistry, University of Surrey, Guildford, United Kingdom
| | - Franca Fraternali
- Randall Centre for Cell & Molecular Biophysics, King’s College London, London, United Kingdom
| | | |
Collapse
|
18
|
The Effect of Waning on Antibody Levels and Memory B Cell Recall following SARS-CoV-2 Infection or Vaccination. Vaccines (Basel) 2022; 10:vaccines10050696. [PMID: 35632452 PMCID: PMC9143792 DOI: 10.3390/vaccines10050696] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
In order to longitudinally track SARS-CoV-2 antibody levels after vaccination or infection, we assessed anti-RBD antibody levels in over 1000 people and found no significant decrease in antibody levels during the first 14 months after infection in unvaccinated participants, however, a significant waning of antibody levels was observed following vaccination. Participants who were pre-immune to SARS-CoV-2 prior to vaccination seroconverted to higher antibody levels, which were maintained at higher levels than in previously infected, unvaccinated participants. Older participants exhibited lower level of antibodies after vaccination, but a higher level after infection than younger people. The rate of antibody waning was not affected by pre-immunity or age. Participants who received a third dose of an mRNA vaccine not only increased their antibody levels ~14-fold, but also had ~3 times more antibodies compared to when they received their primary vaccine series. PBMC-derived memory B cells from 13 participants who lost all circulating antibodies were differentiated into antibody secreting cells (ASCs). There was a significant recall of memory B cell ASCs in the absence of serum antibodies in 5–8 of the 10 vaccinated participants, but not in any of the 3 infected participants, suggesting a strong connection between antibody levels and the effectiveness of memory B cell recall.
Collapse
|
19
|
Forgacs D, Moraes VS, Hanley HB, Gattiker JL, Jefferson AM, Ross TM. The effect of waning on antibody levels and memory B cell recall following SARS-CoV-2 infection or vaccination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.03.16.484099. [PMID: 35313572 PMCID: PMC8936119 DOI: 10.1101/2022.03.16.484099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As of March 2022, there have been over 450 million reported SARS-CoV-2 cases worldwide, and more than 4 billion people have received their primary series of a COVID-19 vaccine. In order to longitudinally track SARS-CoV-2 antibody levels in people after vaccination or infection, a large-scale COVID-19 sero-surveillance progam entitled SPARTA (SeroPrevalence and Respiratory Tract Assessment) was established early in the pandemic. Anti-RBD antibody levels were tracked in more than 1,000 people. There was no significant decrease in antibody levels during the first 14 months after infection in unvaccinated participants, however, significant waning of antibody levels was observed following vaccination, regardless of previous infection status. Moreover, participants who were pre-immune to SARS-CoV-2 prior to vaccination seroconverted to significantly higher antibody levels, and antibodies were maintained at significantly higher levels than in previously infected, unvaccinated participants. This pattern was entirely due to differences in the magnitude of the initial seroconversion event, and the rate of antibody waning was not significantly different based on the pre-immune status. Participants who received a third (booster) dose of an mRNA vaccine not only increased their anti-RBD antibody levels ∼14-fold, but they also had ∼3 times more anti-RBD antibodies compared to the peak of their antibody levels after receiving their primary vaccine series. In order to ascertain whether the presence of serum antibodies is important for long-term seroprotection, PBMCs from 13 participants who lost all detectable circulating antibodies after vaccination or infection were differentiated into memory cells in vitro . There was a significant recall of memory B cells in the absence of serum antibodies in 70% of the vaccinated participants, but not in any of the infected participants. Therefore, there is a strong connection between anti-RBD antibody levels and the effectiveness of memory B cell recall.
Collapse
Affiliation(s)
- David Forgacs
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Vanessa S. Moraes
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Hannah B. Hanley
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Jasper L. Gattiker
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | | | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| |
Collapse
|
20
|
Smit WL, van Tol S, van der Wal S, van Vulpen F, la Grouw S, van Lelyveld L, Limonard G, Bossink A, Godeke GJ, Shrestha S, Reimerink J, Eggink D, Reusken C, Heron M, Thijsen S. Heterologous Immune Responses of Serum IgG and Secretory IgA Against the Spike Protein of Endemic Coronaviruses During Severe COVID-19. Front Immunol 2022; 13:839367. [PMID: 35355988 PMCID: PMC8959642 DOI: 10.3389/fimmu.2022.839367] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/11/2022] [Indexed: 01/08/2023] Open
Abstract
Defining immune correlates of disease severity is important to better understand the immunopathogenesis in COVID-19. Here we made use of a protein microarray platform to detect IgG- and IgA-reactive antibodies in sera and saliva respectively, and assess cross-reactivity between SARS-CoV-2 and endemic coronaviruses (eCoVs). IgG responses against the full protein of spike, but not the S1 subunit, were significantly higher in convalescent sera of patients with severe disease compared to mild disease and healthy controls. In addition, we detected reactivity of secretory IgA to eCoVs in saliva of patients with severe disease, not present in patients with moderate disease or seropositive healthy controls. These heterologous immune responses are in line with non-protective cross-reactivity, and support a potential role for immune imprinting in the pathogenesis of severe COVID-19.
Collapse
Affiliation(s)
- Wouter L. Smit
- Department of Medical Microbiology and Immunology, Diakonessenhuis Utrecht, Utrecht, Netherlands
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sophie van Tol
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Sanne van der Wal
- Department of Medical Microbiology and Immunology, Diakonessenhuis Utrecht, Utrecht, Netherlands
| | - Femke van Vulpen
- Department of Medical Microbiology and Immunology, Diakonessenhuis Utrecht, Utrecht, Netherlands
| | - Shannon la Grouw
- Department of Medical Microbiology and Immunology, Diakonessenhuis Utrecht, Utrecht, Netherlands
| | | | - Gijs Limonard
- Department of Pulmonary Diseases, Diakonessenhuis Utrecht, Utrecht, Netherlands
| | - Ailko Bossink
- Department of Pulmonary Diseases, Diakonessenhuis Utrecht, Utrecht, Netherlands
| | - Gert-Jan Godeke
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Sandhya Shrestha
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Johan Reimerink
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Dirk Eggink
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Chantal Reusken
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Michiel Heron
- Department of Medical Microbiology and Immunology, Diakonessenhuis Utrecht, Utrecht, Netherlands
| | - Steven Thijsen
- Department of Medical Microbiology and Immunology, Diakonessenhuis Utrecht, Utrecht, Netherlands
| |
Collapse
|
21
|
Yu ED, Narowski TM, Wang E, Garrigan E, Mateus J, Frazier A, Weiskopf D, Grifoni A, Premkumar L, da Silva Antunes R, Sette A. Immunological memory to Common Cold Coronaviruses assessed longitudinally over a three-year period. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.03.01.482548. [PMID: 35262082 PMCID: PMC8902883 DOI: 10.1101/2022.03.01.482548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Understanding immune memory to Common Cold Coronaviruses (CCCs) is relevant for assessing its potential impact on the outcomes of SARS-CoV-2 infection, and for the prospects of pan-corona vaccines development. We performed a longitudinal analysis, of pre-pandemic samples collected from 2016-2019. CD4+ T cells and antibody responses specific for CCC and to other respiratory viruses, and chronic or ubiquitous pathogens were assessed. CCC-specific memory CD4+ T cells were detected in most subjects, and their frequencies were comparable to those for other common antigens. Notably, responses to CCC and other antigens such as influenza and Tetanus Toxoid (TT) were sustained over time. CCC-specific CD4+ T cell responses were also associated with low numbers of HLA-DR+CD38+ cells and their magnitude did not correlate with yearly changes in the prevalence of CCC infections. Similarly, spike RBD-specific IgG responses for CCC were stable throughout the sampling period. Finally, high CD4+ T cell reactivity to CCC, but not antibody responses, was associated with high pre-existing SARS-CoV-2 immunity. Overall, these results suggest that the steady and sustained CCC responses observed in the study cohort are likely due to a relatively stable pool of CCC-specific memory CD4+ T cells instead of fast decaying responses and frequent reinfections.
Collapse
Affiliation(s)
- Esther Dawen Yu
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Tara M. Narowski
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7290, USA
| | - Eric Wang
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Emily Garrigan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Jose Mateus
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - April Frazier
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7290, USA
| | - Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| |
Collapse
|
22
|
Jeffery-Smith A, Burton AR, Lens S, Rees-Spear C, Davies J, Patel M, Gopal R, Muir L, Aiano F, Doores KJ, Chow JY, Ladhani SN, Zambon M, McCoy LE, Maini MK. SARS-CoV-2-specific memory B cells can persist in the elderly who have lost detectable neutralizing antibodies. J Clin Invest 2022; 132:e152042. [PMID: 34843448 PMCID: PMC8759779 DOI: 10.1172/jci152042] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 11/24/2021] [Indexed: 11/24/2022] Open
Abstract
Memory B cells (MBCs) can provide a recall response able to supplement waning antibodies (Abs) with an affinity-matured response better able to neutralize variant viruses. We studied a cohort of elderly care home residents and younger staff (median age of 87 years and 56 years, respectively), who had survived COVID-19 outbreaks with only mild or asymptomatic infection. The cohort was selected because of its high proportion of individuals who had lost neutralizing antibodies (nAbs), thus allowing us to specifically investigate the reserve immunity from SARS-CoV-2-specific MBCs in this setting. Class-switched spike and receptor-binding domain (RBD) tetramer-binding MBCs persisted 5 months after mild or asymptomatic SARS-CoV-2 infection, irrespective of age. The majority of spike- and RBD-specific MBCs had a classical phenotype, but we found that activated MBCs, indicating possible ongoing antigenic stimulation or inflammation, were expanded in the elderly group. Spike- and RBD-specific MBCs remained detectable in the majority of individuals who had lost nAbs, although at lower frequencies and with a reduced IgG/IgA isotype ratio. Functional spike-, S1 subunit of the spike protein- (S1-), and RBD-specific recall was also detectable by enzyme-linked immune absorbent spot (ELISPOT) assay in some individuals who had lost nAbs, but was significantly impaired in the elderly. Our findings demonstrate that a reserve of SARS-CoV-2-specific MBCs persists beyond the loss of nAbs but highlight the need for careful monitoring of functional defects in spike- and RBD-specific B cell immunity in the elderly.
Collapse
Affiliation(s)
- Anna Jeffery-Smith
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
- Virus Reference Department, Public Health England (now called UK Health Security Agency [UKHSA]), London, United Kingdom
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Alice R. Burton
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Sabela Lens
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Chloe Rees-Spear
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Jessica Davies
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Monika Patel
- Virus Reference Department, Public Health England (now called UK Health Security Agency [UKHSA]), London, United Kingdom
| | - Robin Gopal
- Virus Reference Department, Public Health England (now called UK Health Security Agency [UKHSA]), London, United Kingdom
| | - Luke Muir
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Felicity Aiano
- Immunisation and Countermeasures Division, Public Health England (now called UKHSA), London, United Kingdom
| | - Katie J. Doores
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - J. Yimmy Chow
- London Coronavirus Response Cell, Public Health England (now called UKHSA), London, United Kingdom
| | - Shamez N. Ladhani
- Immunisation and Countermeasures Division, Public Health England (now called UKHSA), London, United Kingdom
| | - Maria Zambon
- Virus Reference Department, Public Health England (now called UK Health Security Agency [UKHSA]), London, United Kingdom
| | - Laura E. McCoy
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Mala K. Maini
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| |
Collapse
|
23
|
Challenger JD, Foo CY, Wu Y, Yan AWC, Marjaneh MM, Liew F, Thwaites RS, Okell LC, Cunnington AJ. Modelling upper respiratory viral load dynamics of SARS-CoV-2. BMC Med 2022; 20:25. [PMID: 35022051 PMCID: PMC8755404 DOI: 10.1186/s12916-021-02220-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 12/15/2021] [Indexed: 02/09/2023] Open
Abstract
Relationships between viral load, severity of illness, and transmissibility of virus are fundamental to understanding pathogenesis and devising better therapeutic and prevention strategies for COVID-19. Here we present within-host modelling of viral load dynamics observed in the upper respiratory tract (URT), drawing upon 2172 serial measurements from 605 subjects, collected from 17 different studies. We developed a mechanistic model to describe viral load dynamics and host response and contrast this with simpler mixed-effects regression analysis of peak viral load and its subsequent decline. We observed wide variation in URT viral load between individuals, over 5 orders of magnitude, at any given point in time since symptom onset. This variation was not explained by age, sex, or severity of illness, and these variables were not associated with the modelled early or late phases of immune-mediated control of viral load. We explored the application of the mechanistic model to identify measured immune responses associated with the control of the viral load. Neutralising antibodies correlated strongly with modelled immune-mediated control of viral load amongst subjects who produced neutralising antibodies. Our models can be used to identify host and viral factors which control URT viral load dynamics, informing future treatment and transmission blocking interventions.
Collapse
Affiliation(s)
- Joseph D Challenger
- Medical Research Council Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK.
| | - Cher Y Foo
- School of Medicine, Imperial College London, London, UK
| | - Yue Wu
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Ada W C Yan
- Department of Infectious Disease, Imperial College London, London, UK
| | - Mahdi Moradi Marjaneh
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| | - Felicity Liew
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Lucy C Okell
- Medical Research Council Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Aubrey J Cunnington
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK.,Centre for Paediatrics and Child Health, Imperial College London, London, UK
| |
Collapse
|
24
|
Scheiblauer H, Nübling CM, Wolf T, Khodamoradi Y, Bellinghausen C, Sonntagbauer M, Esser-Nobis K, Filomena A, Mahler V, Maier TJ, Stephan C. Antibody response to SARS-CoV-2 for more than one year - kinetics and persistence of detection are predominantly determined by avidity progression and test design. J Clin Virol 2022; 146:105052. [PMID: 34920374 PMCID: PMC8642248 DOI: 10.1016/j.jcv.2021.105052] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/23/2021] [Accepted: 12/02/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Antibody detection of SARS-CoV-2 requires an understanding of its variation, course, and duration. METHODS Antibody response to SARS-CoV-2 was evaluated over 5-430 days on 828 samples across COVID-19 severity levels, for total antibody (TAb), IgG, IgA, IgM, neutralizing antibody (NAb), antibody avidity, and for receptor-binding-domain (RBD), spike (S), or nucleoprotein (N). Specificity was determined on 676 pre-pandemic samples. RESULTS Sensitivity at 30-60 days post symptom onset (pso) for TAb-S/RBD, TAb-N, IgG-S, IgG-N, IgA-S, IgM-RBD, and NAb was 96.6%, 99.5%, 89.7%, 94.3%, 80.9%, 76.9% and 92.8%, respectively. Follow-up 430 days pso revealed: TAb-S/RBD increased slightly (100.0%); TAb-N decreased slightly (97.1%); IgG-S and IgA-S decreased moderately (81.4%, 65.7%); NAb remained positive (94.3%), slightly decreasing in activity after 300 days; there was correlation with IgG-S (Rs = 0.88) and IgA-S (Rs = 0.71); IgG-N decreased significantly from day 120 (15.7%); IgM-RBD dropped after 30-60 days (22.9%). High antibody avidity developed against S/RBD steadily with time in 94.3% of patients after 430 days. This correlated with persistent antibody detection depending on antibody-binding efficiency of the test design. Severe COVID-19 correlated with earlier and higher antibody response, mild COVID-19 was heterogeneous with a wide range of antibody reactivities. Specificity of the tests was ≥99%, except for IgA (96%). CONCLUSION Sensitivity of anti-SARS-CoV-2 assays was determined by test design, target antigen, antibody avidity, and COVID-19 severity. Sustained antibody detection was mainly determined by avidity progression for RBD and S. Testing by TAb and for S/RBD provided the highest sensitivity and longest detection duration of 14 months so far.
Collapse
Affiliation(s)
| | | | - Timo Wolf
- University Hospital Frankfurt - Department of Infectious Diseases and HIV, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Yascha Khodamoradi
- University Hospital Frankfurt - Department of Infectious Diseases and HIV, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Carla Bellinghausen
- University Hospital Frankfurt - Department of Respiratory Medicine and Allergology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Michael Sonntagbauer
- University Hospital Frankfurt - Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | - Angela Filomena
- Paul-Ehrlich-Institute, IVD Testing Laboratory, Langen, Germany
| | - Vera Mahler
- Paul-Ehrlich-Institute, Division Allergology, Langen, Germany
| | - Thorsten Jürgen Maier
- Paul-Ehrlich-Institute, Division Safety of Medicinal Products and Medical Devices, Langen, Germany
| | - Christoph Stephan
- University Hospital Frankfurt - Department of Infectious Diseases and HIV, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
25
|
Abstract
Viruses are essentially, obligate intracellular parasites. They require a host to replicate their genetic material, spread to other cells, and eventually to other hosts. For humans, most viral infections are not considered lethal, regardless if at the cellular level, the virus can obliterate individual cells. Constant genomic mutations, (which can alter the antigenic content of viruses such as influenza or coronaviruses), zoonosis or immunosuppression/immunocompromisation, is when viruses achieve higher host mortality. Frequent examples of the severe consequenses of viral infection can be seen in children and the elderly. In most instances, the immune system will take a multifaceted approach in defending the host against viruses. Depending on the virus, the individual, and the point of entry, the immune system will initiate a robust response which involves multiple components. In this chapter, we expand on the total immune system, breaking it down to the two principal types: Innate and Adaptive Immunity, their different roles in viral recognition and clearance. Finally, how different viruses activate and evade different arms of the immune system.
Collapse
|
26
|
Krashias G, Deeba E, Constantinou A, Hadjiagapiou M, Koptides D, Richter J, Tryfonos C, Bashiardes S, Lambrianides A, Loizidou MA, Hadjisavvas A, Panayiotidis MI, Christodoulou C. Characterization of IgG Antibody Response against SARS-CoV-2 (COVID-19) in the Cypriot Population. Microorganisms 2021; 10:85. [PMID: 35056533 PMCID: PMC8777616 DOI: 10.3390/microorganisms10010085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/16/2021] [Accepted: 12/30/2021] [Indexed: 12/13/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has hit its second year and continues to damage lives and livelihoods across the globe. There continues to be a global effort to present serological data on SARS-CoV-2 antibodies in different individuals. As such, this study aimed to characterize the seroprevalence of SARS-CoV-2 antibodies in the Cypriot population for the first time since the pandemic started. Our results show that a majority of people infected with SARS-CoV-2 developed IgG antibodies against the virus, whether anti-NP, anti-S1RBD, or both, at least 20 days after their infection. Additionally, the percentage of people with at least one antibody against SARS-CoV-2 in the group of volunteers deemed SARS-CoV-2 negative via RT-PCR or who remain untested/undetermined (14.43%) is comparable to other reported percentages worldwide, ranging anywhere from 0.2% to 24%. We postulate that these percentages reflect the underreporting of true infections in the population, and also show the steady increase of herd immunity. Additionally, we showed a significantly marked decrease in anti-NP IgG antibodies in contrast to relatively stable levels of anti-S1RBD IgG antibodies in previously infected individuals across time.
Collapse
Affiliation(s)
- George Krashias
- Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.H.); (A.L.); (M.A.L.); (A.H.); (M.I.P.); (C.C.)
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (E.D.); (A.C.); (D.K.); (J.R.); (C.T.); (S.B.)
| | - Elie Deeba
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (E.D.); (A.C.); (D.K.); (J.R.); (C.T.); (S.B.)
| | - Astero Constantinou
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (E.D.); (A.C.); (D.K.); (J.R.); (C.T.); (S.B.)
| | - Maria Hadjiagapiou
- Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.H.); (A.L.); (M.A.L.); (A.H.); (M.I.P.); (C.C.)
- Department of Neuroimmunology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus
| | - Dana Koptides
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (E.D.); (A.C.); (D.K.); (J.R.); (C.T.); (S.B.)
| | - Jan Richter
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (E.D.); (A.C.); (D.K.); (J.R.); (C.T.); (S.B.)
| | - Christina Tryfonos
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (E.D.); (A.C.); (D.K.); (J.R.); (C.T.); (S.B.)
| | - Stavros Bashiardes
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (E.D.); (A.C.); (D.K.); (J.R.); (C.T.); (S.B.)
| | - Anastasia Lambrianides
- Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.H.); (A.L.); (M.A.L.); (A.H.); (M.I.P.); (C.C.)
- Department of Neuroimmunology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus
| | - Maria A. Loizidou
- Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.H.); (A.L.); (M.A.L.); (A.H.); (M.I.P.); (C.C.)
- Department of Cancer Genetics, Therapeutics and Ultrastructural Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus
| | - Andreas Hadjisavvas
- Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.H.); (A.L.); (M.A.L.); (A.H.); (M.I.P.); (C.C.)
- Department of Cancer Genetics, Therapeutics and Ultrastructural Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus
| | - Mihalis I. Panayiotidis
- Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.H.); (A.L.); (M.A.L.); (A.H.); (M.I.P.); (C.C.)
- Department of Cancer Genetics, Therapeutics and Ultrastructural Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus
| | - Christina Christodoulou
- Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus; (M.H.); (A.L.); (M.A.L.); (A.H.); (M.I.P.); (C.C.)
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (E.D.); (A.C.); (D.K.); (J.R.); (C.T.); (S.B.)
| |
Collapse
|
27
|
Siggins MK, Sriskandan S. Bacterial Lymphatic Metastasis in Infection and Immunity. Cells 2021; 11:33. [PMID: 35011595 PMCID: PMC8750085 DOI: 10.3390/cells11010033] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
Lymphatic vessels permeate tissues around the body, returning fluid from interstitial spaces back to the blood after passage through the lymph nodes, which are important sites for adaptive responses to all types of pathogens. Involvement of the lymphatics in the pathogenesis of bacterial infections is not well studied. Despite offering an obvious conduit for pathogen spread, the lymphatic system has long been regarded to bar the onward progression of most bacteria. There is little direct data on live virulent bacteria, instead understanding is largely inferred from studies investigating immune responses to viruses or antigens in lymph nodes. Recently, we have demonstrated that extracellular bacterial lymphatic metastasis of virulent strains of Streptococcus pyogenes drives systemic infection. Accordingly, it is timely to reconsider the role of lymph nodes as absolute barriers to bacterial dissemination in the lymphatics. Here, we summarise the routes and mechanisms by which an increasing variety of bacteria are acknowledged to transit through the lymphatic system, including those that do not necessarily require internalisation by host cells. We discuss the anatomy of the lymphatics and other factors that influence bacterial dissemination, as well as the consequences of underappreciated bacterial lymphatic metastasis on disease and immunity.
Collapse
Affiliation(s)
- Matthew K. Siggins
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
| | - Shiranee Sriskandan
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2DD, UK
| |
Collapse
|
28
|
Abrokwa SK, Müller SA, Méndez-Brito A, Hanefeld J, El Bcheraoui C. Recurrent SARS-CoV-2 infections and their potential risk to public health - a systematic review. PLoS One 2021; 16:e0261221. [PMID: 34882750 PMCID: PMC8659325 DOI: 10.1371/journal.pone.0261221] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 11/27/2021] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE To inform quarantine and contact-tracing policies concerning re-positive cases-cases testing positive among those recovered. MATERIALS AND METHODS We systematically reviewed and appraised relevant literature from PubMed and Embase for the extent of re-positive cases and their epidemiological characteristics. RESULTS In 90 case reports/series, a total of 276 re-positive cases were found. Among confirmed reinfections, 50% occurred within 90 days from recovery. Four reports related onward transmission. In thirty-five observational studies, rate of re-positives ranged from zero to 50% with no onward transmissions reported. In eight reviews, pooled recurrence rate ranged from 12% to 17.7%. Probability of re-positive increased with several factors. CONCLUSION Recurrence of a positive SARS-CoV-2 test is commonly reported within the first weeks following recovery from a first infection.
Collapse
Affiliation(s)
- Seth Kofi Abrokwa
- Evidence- based Public Health, Centre for International Health Protection, Robert Koch Institute, Berlin, Germany
| | - Sophie Alice Müller
- Centre for International Health Protection, Robert Koch Institute, Berlin, Germany
| | - Alba Méndez-Brito
- Evidence- based Public Health, Centre for International Health Protection, Robert Koch Institute, Berlin, Germany
| | - Johanna Hanefeld
- Centre for International Health Protection, Robert Koch Institute, Berlin, Germany
| | - Charbel El Bcheraoui
- Evidence- based Public Health, Centre for International Health Protection, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
29
|
Does infection with or vaccination against SARS-CoV-2 lead to lasting immunity? THE LANCET. RESPIRATORY MEDICINE 2021; 9:1450-1466. [PMID: 34688434 PMCID: PMC8530467 DOI: 10.1016/s2213-2600(21)00407-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/26/2021] [Accepted: 08/21/2021] [Indexed: 12/17/2022]
Abstract
Many nations are pursuing the rollout of SARS-CoV-2 vaccines as an exit strategy from unprecedented COVID-19-related restrictions. However, the success of this strategy relies critically on the duration of protective immunity resulting from both natural infection and vaccination. SARS-CoV-2 infection elicits an adaptive immune response against a large breadth of viral epitopes, although the duration of the response varies with age and disease severity. Current evidence from case studies and large observational studies suggests that, consistent with research on other common respiratory viruses, a protective immunological response lasts for approximately 5-12 months from primary infection, with reinfection being more likely given an insufficiently robust primary humoral response. Markers of humoral and cell-mediated immune memory can persist over many months, and might help to mitigate against severe disease upon reinfection. Emerging data, including evidence of breakthrough infections, suggest that vaccine effectiveness might be reduced significantly against emerging variants of concern, and hence secondary vaccines will need to be developed to maintain population-level protective immunity. Nonetheless, other interventions will also be required, with further outbreaks likely to occur due to antigenic drift, selective pressures for novel variants, and global population mobility.
Collapse
|
30
|
Rahimi F, Bezmin Abadi AT. The third booster vaccination dose against COVID-19: indication for circulating SARS-CoV-2 variants. Future Virol 2021. [PMID: 34777554 PMCID: PMC8577719 DOI: 10.2217/fvl-2021-0240] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 10/19/2021] [Indexed: 12/15/2022]
Affiliation(s)
- Farid Rahimi
- Research School of Biology, The Australian National University, Canberra, Australia
| | - Amin Talebi Bezmin Abadi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
31
|
Tian X, Jiang W, Zhang H, Lu X, Li L, Liu W, Li J. Persistence of the SARS-CoV-2 Antibody Response in Asymptomatic Patients in Correctional Facilities. Front Microbiol 2021; 12:789374. [PMID: 34858383 PMCID: PMC8631518 DOI: 10.3389/fmicb.2021.789374] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/21/2021] [Indexed: 12/23/2022] Open
Abstract
SARS-CoV-2 has caused a global health disaster with millions of death worldwide, and the substantial proportion of asymptomatic carriers poses a huge threat to public health. The long-term antibody responses and neutralization activity during natural asymptomatic SARS-CoV-2 infection are unknown. In this study, we used enzyme-linked immunosorbent assays (ELISA) and neutralization assay with purified SARS-CoV-2S and N proteins to study the antibody responses of 156 individuals with natural asymptomatic infection. We found robust antibody responses to SARS-CoV-2 in 156 patients from 6 to 12 months. Although the antibody responses gradually decreased, S-IgG was more stable than N-IgG. S-IgG was still detected in 79% of naturally infected individuals after 12 months of infection. Moderate to potent neutralization activities were also observed in 98.74% of patients 6 months after infection. However, this proportion decreased at 8-month (46.15%) and 10-month (39.11%) after infection, respectively. Only 23.72% of patients displayed potent neutralization activity at 12 months. This study strongly supports the long-term presence of antibodies against SARS-CoV-2 in individuals with natural asymptomatic infection, although the magnitude of the antibody responses started to cripple 6 months after infection.
Collapse
Affiliation(s)
- Xiaodong Tian
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Wenguo Jiang
- Jining Center for Disease Control and Prevention, Shandong, China
| | - He Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - XiXi Lu
- Jining Center for Disease Control and Prevention, Shandong, China
| | - Libo Li
- Jining Center for Disease Control and Prevention, Shandong, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Savaid Medical School, University of the Chinese Academy of Sciences, Beijing, China
- Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China
| | - Jing Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of the Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
32
|
Heinzel C, Pinilla YT, Elsner K, Friessinger E, Mordmüller B, Kremsner PG, Held J, Fendel R, Kreidenweiss A. Non-Invasive Antibody Assessment in Saliva to Determine SARS-CoV-2 Exposure in Young Children. Front Immunol 2021; 12:753435. [PMID: 34691072 PMCID: PMC8531807 DOI: 10.3389/fimmu.2021.753435] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/22/2021] [Indexed: 11/13/2022] Open
Abstract
Saliva is a body fluid with hitherto unused potential for the assessment of SARS-CoV-2 antibodies. Specific antibodies can indicate a past SARS-CoV-2 infection and allow to estimate the proportion of individuals with a potential protective immunity. First, we carefully characterized plasma samples obtained from adult control groups with and without prior SARS-CoV-2 infection using certified reference ELISAs. Simultaneously collected saliva samples of confirmed convalescent and negative individuals where then used to validate the herein newly developed ELISA for the detection of SARS-CoV-2 IgG antibodies in saliva. The saliva ELISA was applied to assess SARS-CoV-2 exposure in young children (N = 837) in the age between 1 and 10 years in Tübingen, Germany, towards the end of the first pandemic year 2020. Sensitivity and specificity of the new saliva ELISA was 87% and 100%, respectively. With 12% of all Tübingen children sampled via their respective educational institutions, estimates of SARS-CoV-2 antibody prevalence was 1.6%. Interestingly, only 0.4% preschool kids were positive compared to 3.0% of primary school children. Less than 20% of positive children self-reported symptoms within two months prior to saliva sampling that could be associated - but not exclusively - with a SARS-CoV-2 infection. The saliva ELISA is a valid and suitable protocol to enable population-based surveys for SARS-CoV-2 antibodies. Using non-invasive sampling and saliva ELISA testing, we found that prevalence of SARS-CoV-2 antibodies was significantly lower in young children than in primary school children.
Collapse
Affiliation(s)
- Constanze Heinzel
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Yudi T Pinilla
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Käthe Elsner
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Evelyn Friessinger
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Benjamin Mordmüller
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter G Kremsner
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Jana Held
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Rolf Fendel
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Andrea Kreidenweiss
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| |
Collapse
|