1
|
Meyer J, Nadal J, Batsa Debrah L, Debrah AY, Osei-Mensah J, Adu Mensah D, Korir PJ, Kuehlwein JM, Klarmann-Schulz U, Hoerauf A, Adjobimey T. Robust COVID-19 Vaccine Responses Despite Filarial Co-Infection: Insights from a Lymphatic Filariasis Cohort in Ghana. Vaccines (Basel) 2025; 13:312. [PMID: 40266230 PMCID: PMC11945955 DOI: 10.3390/vaccines13030312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND/OBJECTIVES Although the COVID-19 pandemic has largely concluded, the varied trajectories it has followed in different regions of the world remain incompletely understood. Intensive research is needed to fully grasp its course and the implications for future global health challenges. Notably, the milder trajectory of the COVID-19 pandemic in Sub-Saharan Africa has defied initial predictions. An emerging body of evidence suggests that, in addition to the continent's younger average age and the lower prevalence of relevant comorbidities, co-infections with helminths may have also impressively shaped the pandemic's milder trajectory in the region. Indeed, helminths are renowned for their ability to modulate human immune responses, which, while potentially beneficial in limiting excessive inflammation, could also diminish vaccine efficacy and impede viral clearance. This study investigated different aspects of the intricate interactions between COVID-19 and Lymphatic Filariasis (LF), a helminth infection caused by parasitic worms such as Wuchereria bancrofti, Brugia malayi, and Brugia timori and endemic to various regions in Sub-Saharan Africa and the tropics. METHODS For this purpose, samples of a larger and ongoing clinical trial (ethical approval codes: CHRPE/AP/525/17 and 325/21; trial registration number ISRCTN14042737) were collected from 222 individuals from endemic areas of Ghana, along with comprehensive clinical and demographic data. The samples include LF patients (n = 222) grouped according to their Lymphoedema (LE) stages, as well as COVID-19 vaccinated (n = 81) and non-vaccinated individuals (n = 141). All vaccinated participants received the COVID-19 vaccine ChAdOx1-S (also known as Vaxzevria) developed by the University of Oxford and AstraZenca. The expressions of SARS-CoV-2 and filarial-specific antibodies (IgG, IgA) were accessed using ELISA, while Luminex-based immunoassays were employed to measure the expression of SARS-CoV-2 variant-specific neutralizing antibodies. The interplay between vaccine responses and demographic factors was analyzed using group comparisons with the Kruskal-Wallis or Mann-Whitney U tests. RESULTS The results indicate that a remarkable portion of unvaccinated individuals (56% IgA seropositive, 39% IgG seropositive) developed antibodies against SARS-CoV-2 despite no confirmed infection. Notably, the study identified a robust antibody response to COVID-19 vaccination, which was independent of the degree of LF pathology or parasitic status. An important observation was the reduced SARS-CoV-2 antibody response in individuals seropositive for Ascaris lumbricoides (p = 0.0264), highlighting an interaction between roundworm infection and COVID-19. CONCLUSIONS The study concludes that the ChAdOx1-S COVID-19 vaccine (AstraZeneca) triggers a strong immune response in LF patients; however, filarial and/or soil-transmitted helminth seropositivity might influence the COVID-19 infection-induced response. These findings emphasize the complexity of infectious disease dynamics in co-infected populations and the need to decipher parasite-induced immunomodulatory mechanisms on COVID-19 vaccination.
Collapse
Affiliation(s)
- Julia Meyer
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany
| | - Jennifer Nadal
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany
- Institute for Medical Biometry, Informatics, and Epidemiology (IMBIE), University Hospital Bonn, 53127 Bonn, Germany
| | - Linda Batsa Debrah
- Department of Clinical Microbiology, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi 00233, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology (KNUST), Kumasi 00233, Ghana
| | - Alexander Yaw Debrah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology (KNUST), Kumasi 00233, Ghana
- Faculty of Allied Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi 00233, Ghana
| | - Jubin Osei-Mensah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology (KNUST), Kumasi 00233, Ghana
- Department of Pathobiology, School of Veterinary Medicine, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi 00233, Ghana
| | - Derrick Adu Mensah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology (KNUST), Kumasi 00233, Ghana
- Department of Medical Laboratory Technology, Royal Ann College of Health, Kumasi 00233, Ghana
| | - Patricia Jebett Korir
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany
- Bonn-Cologne Site, German Center for Infectious Disease Research (DZIF), 53113 Bonn, Germany
| | - Janina M. Kuehlwein
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany
- Bonn-Cologne Site, German Center for Infectious Disease Research (DZIF), 53113 Bonn, Germany
| | - Ute Klarmann-Schulz
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany
- Bonn-Cologne Site, German Center for Infectious Disease Research (DZIF), 53113 Bonn, Germany
| | - Achim Hoerauf
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany
- Bonn-Cologne Site, German Center for Infectious Disease Research (DZIF), 53113 Bonn, Germany
| | - Tomabu Adjobimey
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany
- Laboratoire de Biologie Intégrative Pour l’Innovation Thérapeutique (BioInov), Faculté des Sciences et Techniques (FAST), Université d’Abomey Calavi, Abomey Calavi BP 526, Benin
| |
Collapse
|
2
|
Hoffmann DJ, Meyer PWA, Worsley CM, van der Mescht MA, Visser A, Pillay TS. SARS-CoV-2 seropositivity amongst healthcare workers in South Africa during the Omicron wave: natural infection versus vaccination. J Clin Pathol 2025:jcp-2024-209722. [PMID: 39824540 DOI: 10.1136/jcp-2024-209722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 12/09/2024] [Indexed: 01/20/2025]
Abstract
AIMS Concerns over population-level immunity have been heightened with each successive wave of COVID-19, prompting questions about whether it is primarily derived from vaccination efforts or from previous natural infections with the virus. We wished to determine the seroprevalence of SARS-CoV-2 antibodies among healthcare workers (HCWs) in Pretoria (Tshwane), South Africa, and to establish whether they were derived from vaccination or natural infection. METHODS Serum samples were collected from HCWs during the fourth wave of COVID-19 between 1 December 2021 and 13 March 2022. The samples were tested using the Abbott SARS-CoV-2 Spike IgG (S-IgG), IgM (S-IgM) and the SARS-CoV-2 Nucleocapsid IgG (NC-IgG) kits. RESULTS Of the 221 participants, 76% (n=168) were women and 24% (n=53) were men. A total of 96.4% (n=213) of the participants were vaccinated. Natural infection-derived antibodies were detected in 23% (n=51) of participants, and vaccine-derived antibodies in 74% (n=164) of the HCWs. CONCLUSIONS Even after three waves of COVID-19, HCWs derived most of their detectable antibodies from vaccination. Vaccination remains an essential tool to protect HCWs and patients from SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Daniel J Hoffmann
- Department of Clinical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- National Health Laboratory Service, Pretoria, South Africa
| | - Pieter W A Meyer
- National Health Laboratory Service, Pretoria, South Africa
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Catherine M Worsley
- National Health Laboratory Service, Pretoria, South Africa
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Mieke A van der Mescht
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - A Visser
- Department of Clinical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- National Health Laboratory Service, Pretoria, South Africa
| | - Tahir S Pillay
- Department of Clinical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- National Health Laboratory Service, Pretoria, South Africa
- Department of Chemical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
3
|
Dwivedi T, Raj A, Das N, Gupta R, Bhatnagar S, Mohan A, Guleria R. Persistence of SARS-CoV-2 Antibodies for a Year Following SARS-CoV-2 Vaccinations (BBV152 and ChAdOx1 nCoV-19). Indian J Clin Biochem 2025; 40:111-120. [PMID: 39835239 PMCID: PMC11741955 DOI: 10.1007/s12291-023-01149-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/21/2023] [Indexed: 01/22/2025]
Abstract
The first two vaccines administered in the COVID-19 vaccination campaign of India were Covaxin (BBV152) and Covishield (ChAdOx1-nCoV-19). In this study, we evaluate the longevity and sustainability of the humoral immune response after vaccination and various factors influencing it. An observational study was conducted in individuals who received both doses of Covaxin or Covishield vaccine, and their blood samples were analyzed for total-antiRBD-SARS-CoV-2 antibodies. Then, antibody titers were classified based on monthly time-intervals up to 360 days and their trend was analyzed. In addition, the correlation between antibody titers and factors such as previous SARS-CoV-2-infection status, vaccine type and presence of comorbidities was examined. Of the 2069 participants, most (1767;85.4%) had been vaccinated with Covaxin, but the higher antibody titers were induced by Covishield vaccine at all time points. However overall, antibodies persisted for at least 1 year, although a drop in antibody titers occurred in the 3rd and 6th months. In addition, 430 (20.8%) participants had prior SARS-CoV-2 infection (hybrid immunity) with a significantly higher humoral immune response compared with vaccine-induced immunity (naive immunity). No significant differences were observed in antibody titers related to age, sex and presence of comorbidities. We concluded that vaccine-mediated immunity lasts for at least one year. However, antibody titers decrease over time, which may be more pronounced in certain groups such as Covaxin vaccine, vaccine-induced-immunity, presence of comorbidities and > 60 years which should be considered when recommending booster vaccination, as these individuals may have a stronger and longer-lasting immune response to the virus.
Collapse
Affiliation(s)
- Tanima Dwivedi
- Laboratory Oncology, National Cancer Institute, All India Institute of Medical Sciences, Jhajjar, Haryana 124105 India
| | - Apurva Raj
- Laboratory Oncology, National Cancer Institute, All India Institute of Medical Sciences, Jhajjar, Haryana 124105 India
| | - Nupur Das
- Laboratory Oncology, National Cancer Institute, All India Institute of Medical Sciences, Jhajjar, Haryana 124105 India
| | - Ritu Gupta
- Laboratory Oncology, Dr BRAIRCH, All India Institute of Medical Sciences, New Delhi, 110029 India
| | - Sushma Bhatnagar
- Department of Onco-Anesthesiology and Palliative Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Anant Mohan
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Randeep Guleria
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
4
|
Sun L, Zhou B, Guo Y, Huang Y, Tang Z, Wu Z, Li Y, Zhao L, Shan Y, Xu X, Jiang H. Clinical characteristics and booster vaccine effectiveness of the Omicron variant. BMC Infect Dis 2024; 24:1351. [PMID: 39593005 PMCID: PMC11600739 DOI: 10.1186/s12879-024-10261-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Although many people received the COVID-19 vaccine, shortly after the Chinese government announced that the three-year COVID-19 restrictions were being eased, the first large number of Omicron infections appeared in Beijing. We describe epidemiological characteristics, clinical severity, and time-to-event distribution of patients infected with SARS-CoV-2 in Beijing Omicron outbreak, comparing those who received the booster vaccine with cases of full-course/partial-course vaccines. METHODS We collected epidemiological, clinical, laboratory, and clinical management data from the hospital information system (HIS) for 1495 cases during Dec 2022-Jan 2023. We also collected illness onset time, diagnosis time, hospital admission time, and start and end times of each treatment. In addition, we collected the time of vaccination, inoculation times, and type of COVID-19 vaccination thorough the vaccination system. We described the epidemiological characteristics across vaccine inoculation doses, and estimated the risk of death, mechanical ventilation, and admission to the intensive care unit for patients admitted to hospital. We used the Kaplan-Meier method to estimate the survival rate and plot the survival curve, and the Cox proportional hazards model to assess the effect of covariates on survival time. RESULTS Of the 1495 cases, 58.1% were male. The median ages in the non-vaccinated and vaccinated groups were 80 and 47 years. Elderly with underlying medical conditions and lower BMI less willing to vaccinate(p < 0.05). Both the inactivated vaccine and adenovirus vaccine could reduce clinical severity and prolong survival time, and the protective effect of booster vaccination was the best. The clinical severity increased linearly from the booster vaccinated group to the full-course vaccinated group and non-vaccinated groups, and the death risk of COVID-19 cases without vaccination was the highest. CONCLUSION Booster vaccines of COVID-19 can provide greater protection against severe illness and death, and continuous monitoring and regular assessment are needed to minimize the risk of a recurrence of the pandemic.
Collapse
Affiliation(s)
- Lijun Sun
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Biye Zhou
- The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China
| | - Yijia Guo
- Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Yunfei Huang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zihui Tang
- The North Campus of the Fifth Medical Center of the General Hospital of the People's Liberation Army, Beijing, 100039, China
| | - Zhaojun Wu
- Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Yuwei Li
- Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Liping Zhao
- Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Yi Shan
- The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China
| | - Xiaolong Xu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Hui Jiang
- Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
- Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| |
Collapse
|
5
|
Kadelka S, Bouman JA, Ashcroft P, Regoes RR. Correcting for Antibody Waning in Cumulative Incidence Estimation From Sequential Serosurveys. Am J Epidemiol 2024; 193:777-786. [PMID: 38012125 PMCID: PMC11074712 DOI: 10.1093/aje/kwad226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 08/31/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023] Open
Abstract
Serosurveys are a widely used tool to estimate the cumulative incidence-the fraction of a population that has been infected by a given pathogen. These surveys rely on serological assays that measure the level of pathogen-specific antibodies. Because antibody levels are waning, the fraction of previously infected individuals that have seroreverted increases with time past infection. To avoid underestimating the true cumulative incidence, it is therefore essential to correct for waning antibody levels. We present an empirically supported approach for seroreversion correction in cumulative incidence estimation when sequential serosurveys are conducted in the context of a newly emerging infectious disease. The correction is based on the observed dynamics of antibody titers in seropositive cases and validated using several in silico test scenarios. Furthermore, through this approach we revise a previous cumulative incidence estimate relying on the assumption of an exponentially declining probability of seroreversion over time, of severe acute respiratory syndrome coronavirus 2, of 76% in Manaus, Brazil, by October 2020 to 47.6% (95% confidence region: 43.5-53.5). This estimate has implications, for example, for the proximity to herd immunity in Manaus in late 2020.
Collapse
Affiliation(s)
- Sarah Kadelka
- Correspondence to Dr. Sarah Kadelka, ETH Zürich, Institut für Integrative Biologie, CHN K 12.2, Universitätstrasse 16, 8092 Zürich, Switzerland (e-mail: ); or Prof. Dr. Roland R. Regoes, ETH Zürich, Institut für Integrative Biologie, CHN K 12.2, Universitätstrasse 16, 8092 Zürich, Switzerland (e-mail: )
| | | | | | - Roland R Regoes
- Correspondence to Dr. Sarah Kadelka, ETH Zürich, Institut für Integrative Biologie, CHN K 12.2, Universitätstrasse 16, 8092 Zürich, Switzerland (e-mail: ); or Prof. Dr. Roland R. Regoes, ETH Zürich, Institut für Integrative Biologie, CHN K 12.2, Universitätstrasse 16, 8092 Zürich, Switzerland (e-mail: )
| |
Collapse
|
6
|
Zhang LN, Tan JT, Ng HY, Liao YS, Zhang RQ, Chan KH, Hung IFN, Lam TTY, Cheung KS. Association between Gut Microbiota Composition and Long-Term Vaccine Immunogenicity following Three Doses of CoronaVac. Vaccines (Basel) 2024; 12:365. [PMID: 38675747 PMCID: PMC11055114 DOI: 10.3390/vaccines12040365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Neutralizing antibody level wanes with time after COVID-19 vaccination. We aimed to study the relationship between baseline gut microbiota and immunogenicity after three doses of CoronaVac. METHODS This was a prospective cohort study recruiting three-dose CoronaVac recipients from two centers in Hong Kong. Blood samples were collected at baseline and one year post-first dose for virus microneutralization (vMN) assays to determine neutralization titers. The primary outcome was high immune response (defined as with vMN titer ≥ 40). Shotgun DNA metagenomic sequencing of baseline fecal samples identified potential bacterial species and metabolic pathways using Linear Discriminant Analysis Effect Size (LEfSe) analysis. Univariate and multivariable logistic regression models were used to identify high response predictors. RESULTS In total, 36 subjects were recruited (median age: 52.7 years [IQR: 47.9-56.4]; male: 14 [38.9%]), and 18 had low immune response at one year post-first dose vaccination. Eubacterium rectale (log10LDA score = 4.15, p = 0.001; relative abundance of 1.4% vs. 0, p = 0.002), Collinsella aerofaciens (log10LDA score = 3.31, p = 0.037; 0.39% vs. 0.18%, p = 0.038), and Streptococcus salivarius (log10LDA score = 2.79, p = 0.021; 0.05% vs. 0.02%, p = 0.022) were enriched in low responders. The aOR of high immune response with E. rectale, C. aerofaciens, and S. salivarius was 0.03 (95% CI: 9.56 × 10-4-0.32), 0.03 (95% CI: 4.47 × 10-4-0.59), and 10.19 (95% CI: 0.81-323.88), respectively. S. salivarius had a positive correlation with pathways enriched in high responders like incomplete reductive TCA cycle (log10LDA score = 2.23). C. aerofaciens similarly correlated with amino acid biosynthesis-related pathways. These pathways all showed anti-inflammation functions. CONCLUSION E. rectale,C. aerofaciens, and S. salivarius correlated with poorer long-term immunogenicity following three doses of CoronaVac.
Collapse
Affiliation(s)
- Li-Na Zhang
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Jing-Tong Tan
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Ho-Yu Ng
- School of Clinical Medicine, The University of Hong Kong, Hong Kong
| | - Yun-Shi Liao
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong
- Centre for Immunology & Infection Limited, 17W Hong Kong Science & Technology Parks, Hong Kong
| | - Rui-Qi Zhang
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Kwok-Hung Chan
- Centre for Immunology & Infection Limited, 17W Hong Kong Science & Technology Parks, Hong Kong
| | - Ivan Fan-Ngai Hung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Tommy Tsan-Yuk Lam
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong
| | - Ka-Shing Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| |
Collapse
|
7
|
Abela IA, Schwarzmüller M, Ulyte A, Radtke T, Haile SR, Ammann P, Raineri A, Rueegg S, Epp S, Berger C, Böni J, Manrique A, Audigé A, Huber M, Schreiber PW, Scheier T, Fehr J, Weber J, Rusert P, Günthard HF, Kouyos RD, Puhan MA, Kriemler S, Trkola A, Pasin C. Cross-protective HCoV immunity reduces symptom development during SARS-CoV-2 infection. mBio 2024; 15:e0272223. [PMID: 38270455 PMCID: PMC10865973 DOI: 10.1128/mbio.02722-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/15/2023] [Indexed: 01/26/2024] Open
Abstract
Numerous clinical parameters link to severe coronavirus disease 2019, but factors that prevent symptomatic disease remain unknown. We investigated the impact of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) and endemic human coronavirus (HCoV) antibody responses on symptoms in a longitudinal children cohort (n = 2,917) and a cross-sectional cohort including children and adults (n = 882), all first exposed to SARS-CoV-2 (March 2020 to March 2021) in Switzerland. Saliva (n = 4,993) and plasma (n = 7,486) antibody reactivity to the four HCoVs (subunit S1 [S1]) and SARS-CoV-2 (S1, receptor binding domain, subunit S2 [S2], nucleocapsid protein) was determined along with neutralizing activity against SARS-CoV-2 Wuhan, Alpha, Delta, and Omicron (BA.2) in a subset of individuals. Inferred recent SARS-CoV-2 infection was associated with a strong correlation between mucosal and systemic SARS-CoV-2 anti-spike responses. Individuals with pre-existing HCoV-S1 reactivity exhibited significantly higher antibody responses to SARS-CoV-2 in both plasma (IgG regression coefficients = 0.20, 95% CI = [0.09, 0.32], P < 0.001) and saliva (IgG regression coefficient = 0.60, 95% CI = [0.088, 1.11], P = 0.025). Saliva neutralization activity was modest but surprisingly broad, retaining activity against Wuhan (median NT50 = 32.0, 1Q-3Q = [16.4, 50.2]), Alpha (median NT50 = 34.9, 1Q-3Q = [26.0, 46.6]), and Delta (median NT50 = 28.0, 1Q-3Q = [19.9, 41.7]). In line with a rapid mucosal defense triggered by cross-reactive HCoV immunity, asymptomatic individuals presented with higher pre-existing HCoV-S1 activity in plasma (IgG HKU1, odds ratio [OR] = 0.53, 95% CI = [0.29,0.97], P = 0.038) and saliva (total HCoV, OR = 0.55, 95% CI = [0.33, 0.91], P = 0.019) and higher SARS-CoV-2 reactivity in saliva (IgG S2 fold change = 1.26, 95% CI = [1.03, 1.54], P = 0.030). By investigating the systemic and mucosal immune responses to SARS-CoV-2 and HCoVs in a population without prior exposure to SARS-CoV-2 or vaccination, we identified specific antibody reactivities associated with lack of symptom development.IMPORTANCEKnowledge of the interplay between human coronavirus (HCoV) immunity and severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection is critical to understanding the coexistence of current endemic coronaviruses and to building knowledge potential future zoonotic coronavirus transmissions. This study, which retrospectively analyzed a large cohort of individuals first exposed to SARS-CoV-2 in Switzerland in 2020-2021, revealed several key findings. Pre-existing HCoV immunity, particularly mucosal antibody responses, played a significant role in improving SARS-CoV-2 immune response upon infection and reducing symptoms development. Mucosal neutralizing activity against SARS-CoV-2, although low in magnitude, retained activity against SARS-CoV-2 variants underlining the importance of maintaining local mucosal immunity to SARS-CoV-2. While the cross-protective effect of HCoV immunity was not sufficient to block infection by SARS-CoV-2, the present study revealed a remarkable impact on limiting symptomatic disease. These findings support the feasibility of generating pan-protective coronavirus vaccines by inducing potent mucosal immune responses.
Collapse
Affiliation(s)
- Irene A. Abela
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - Agne Ulyte
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Thomas Radtke
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Sarah R. Haile
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Priska Ammann
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Alessia Raineri
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Sonja Rueegg
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Selina Epp
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | | | - Jürg Böni
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Amapola Manrique
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Annette Audigé
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Michael Huber
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Peter W. Schreiber
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Thomas Scheier
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jan Fehr
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Jacqueline Weber
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Peter Rusert
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Huldrych F. Günthard
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Roger D. Kouyos
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Milo A. Puhan
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Susi Kriemler
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Alexandra Trkola
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Chloé Pasin
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Collegium Helveticum, Zurich, Switzerland
| |
Collapse
|
8
|
Kassianos G, MacDonald P, Aloysius I, Pather S. Responses to Common Misconceptions Relating to COVID-19 Variant-Adapted mRNA Vaccines. Vaccines (Basel) 2024; 12:57. [PMID: 38250870 PMCID: PMC10819631 DOI: 10.3390/vaccines12010057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024] Open
Abstract
The evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the waning of immunity over time has necessitated the use of booster doses of original coronavirus disease 2019 (COVID-19) vaccines. This has also led to the development and implementation of variant-adapted messenger RNA (mRNA) vaccines that include an Omicron sub-lineage component in addition to the antigen based on the wild-type virus spike protein. Subsequent emergence of the recombinant XBB sub-lineages triggered the development of monovalent XBB-based variant-adapted mRNA vaccines, which are available for vaccination campaigns in late 2023. Misconceptions about new variant-adapted vaccines may exacerbate vaccine fatigue and drive the lack of vaccine acceptance. This article aims to address common concerns about the development and use of COVID-19 variant-adapted mRNA vaccines that have emerged as SARS-CoV-2 has continued to evolve.
Collapse
Affiliation(s)
- George Kassianos
- Royal College of General Practitioners, London NW1 2FB, UK;
- British Global and Travel Health Association, London NW1 2FB, UK
| | | | | | | |
Collapse
|
9
|
Eybpoosh S, Biglari A, Sorouri R, Ashrafian F, Sadat Larijani M, Verez-Bencomo V, Toledo-Romani ME, Valenzuela Silva C, Salehi-Vaziri M, Dahmardeh S, Doroud D, Banifazl M, Mostafavi E, Bavand A, Ramezani A. Immunogenicity and safety of heterologous boost immunization with PastoCovac Plus against COVID-19 in ChAdOx1-S or BBIBP-CorV primed individuals. PLoS Pathog 2023; 19:e1011744. [PMID: 37910480 PMCID: PMC10619776 DOI: 10.1371/journal.ppat.1011744] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 10/10/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND This study aimed at evaluation and comparison of PastoCovac Plus protein-subunit vaccine in parallel with ChAdOx1-S (AstraZeneca) and BBIBP-CorV (Sinopharm) in primarily vaccinated volunteers with two doses of ChAdOx1-S or BBIBP-CorV. MATERIALS AND METHODS 194 volunteers enrolled the study who were previously primed with 2 doses of ChAdOx1-S or BBIBP-CorV vaccines. They were divided into two heterologous regimens receiving a third dose of PastoCovac Plus, and two parallel homologous groups receiving the third dose of BBIBP-CorV or ChAdOx1-S. Serum samples were obtained just before and 4 weeks after booster dose. Anti-spike IgG and neutralizing antibodies were quantified and the conventional live-virus neutralization titer, (cVNT50) assay was done against Omicron BA.5 variant. Moreover, the adverse events data were recorded after receiving booster doses. RESULTS ChAdOx1-S/PastoCovac Plus group reached 73.0 units increase in anti-Spike IgG rise compared to the ChAdOx1-S/ ChAdOx1-S (P: 0.016). No significant difference was observed between the two groups regarding neutralizing antibody rise (P: 0.256), indicating equivalency of both booster types. Adjusting for baseline titers, the BBIBP-CorV/PastoCovac Plus group showed 135.2 units increase (P<0.0001) in anti-Spike IgG, and 3.1 (P: 0.008) unit increase in mean rise of neutralizing antibodies compared to the homologous group. Adjustment for COVID-19 history, age, underlying diseases, and baseline antibody titers increased the odds of anti-Spike IgG fourfold rise both in the ChAdOx1-S (OR: 1.9; P: 0.199) and BBIBP CorV (OR: 37.3; P< 0.0001) heterologous groups compared to their corresponding homologous arms. The odds of neutralizing antibody fourfold rise, after adjustment for the same variables, was 2.4 (P: 0.610) for the ChAdOx1-S heterologous group and 5.4 (P: 0.286) for the BBIBP CorV heterologous groups compared to their corresponding homologous groups. All the booster types had the potency to neutralize BA.5 variant with no significant difference. The highest rate of adverse event incidence was recorded for ChAdOx1-S homologous group. CONCLUSIONS PastoCovac Plus booster application in primed individuals with BBIBP-CorV or ChAdOx1-S successfully increased specific antibodies' levels without any serious adverse events. This vaccine could be administrated in the heterologous regimen to effectively boost humoral immune responses.
Collapse
Affiliation(s)
- Sana Eybpoosh
- Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Biglari
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Rahim Sorouri
- IPI Directorate, Pasteur Institute of Iran, Tehran, Iran
- Department of Microbiology, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ashrafian
- Clinical Research Department, Pasteur Institute of Iran, Tehran, Iran
| | | | | | | | | | | | - Sarah Dahmardeh
- Vaccination Department, Pasteur Institute of Iran, Tehran, Iran
| | - Delaram Doroud
- Quality Control Department, Production and research Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Banifazl
- Iranian Society for Support of Patients with Infectious Disease, Tehran, Iran
| | - Ehsan Mostafavi
- Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Anahita Bavand
- Clinical Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Amitis Ramezani
- Clinical Research Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
10
|
Widyasari K, Jang J, Kang T, Kim S. Effectiveness of Bivalent Omicron-Containing Booster Vaccines against SARS-CoV-2 Omicron Variant among Individuals with and without Prior SARS-CoV-2 Infection. Viruses 2023; 15:1756. [PMID: 37632098 PMCID: PMC10459014 DOI: 10.3390/v15081756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
In this study, we evaluated the effectiveness of the bivalent mRNA COVID-19 vaccines against the Omicron variant in individuals with or without prior SARS-CoV-2 infection history. We assessed the SARS-CoV-2-specific neutralizing antibody in serum samples by surrogate virus neutralizing assay (sVNT) and determined the serum's neutralizing capacity against the Omicron BA.5 by a plaque reduction neutralizing test (PRNT50). The results of the sVNT assay demonstrate a higher percentage of inhibition of the serum samples from the infected group than from the uninfected group (p = 0.01) before the bivalent vaccination but a similarly high percentage of inhibition after the vaccination. Furthermore, the results of the PRNT50 assay demonstrate a higher neutralizing capacity of the serum samples against Omicron BA.5 in the infected group compared to the uninfected group, both before and after the bivalent vaccine administration (p < 0.01 and p = 0.02 for samples collected before and after the bivalent vaccination, respectively). A higher neutralizing capacity of the serum samples against BA.5 following bivalent vaccination compared to those before vaccination suggests the efficacy of bivalent mRNA COVID-19 vaccines in triggering an immune response against the Omicron variant, particularly BA.5, regardless of infection history.
Collapse
Affiliation(s)
- Kristin Widyasari
- Gyeongsang Institute of Health Sciences, Gyeongsang National University, Jinju 52727, Republic of Korea;
| | - Jieun Jang
- Gyeongnam Center for Infectious Disease Control and Prevention, Changwon 51154, Republic of Korea;
| | - Taejoon Kang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejon 34141, Republic of Korea;
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sunjoo Kim
- Gyeongsang Institute of Health Sciences, Gyeongsang National University, Jinju 52727, Republic of Korea;
- Gyeongnam Center for Infectious Disease Control and Prevention, Changwon 51154, Republic of Korea;
- Department of Laboratory Medicine, College of Medicine Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Laboratory Medicine, Gyeongsang National University Changwon Hospital, Changwon 51472, Republic of Korea
| |
Collapse
|
11
|
den Hartog Y, Malahe SRK, Rietdijk WJR, Dieterich M, Gommers L, Geers D, Bogers S, van Baarle D, Diavatopoulos DA, Messchendorp AL, van der Molen RG, Remmerswaal EBM, Bemelman FJ, Gansevoort RT, Hilbrands LB, Sanders JS, GeurtsvanKessel CH, Kho MML, Reinders MEJ, de Vries RD, Baan CC. Th 1-dominant cytokine responses in kidney patients after COVID-19 vaccination are associated with poor humoral responses. NPJ Vaccines 2023; 8:70. [PMID: 37198189 DOI: 10.1038/s41541-023-00664-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 04/25/2023] [Indexed: 05/19/2023] Open
Abstract
Cytokines are regulators of the immune response against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). However, the contribution of cytokine-secreting CD4+ and CD8+ memory T cells to the SARS-CoV-2-specific humoral immune response in immunocompromised kidney patients is unknown. Here, we profiled 12 cytokines after stimulation of whole blood obtained 28 days post second 100 μg mRNA-1273 vaccination with peptides covering the SARS-CoV-2 spike (S)-protein from patients with chronic kidney disease (CKD) stage 4/5, on dialysis, kidney transplant recipients (KTR), and healthy controls. Unsupervised hierarchical clustering analysis revealed two distinct vaccine-induced cytokine profiles. The first profile was characterized by high levels of T-helper (Th)1 (IL-2, TNF-α, and IFN-γ) and Th2 (IL-4, IL-5, IL-13) cytokines, and low levels of Th17 (IL-17A, IL-22) and Th9 (IL-9) cytokines. This cluster was dominated by patients with CKD, on dialysis, and healthy controls. In contrast, the second cytokine profile contained predominantly KTRs producing mainly Th1 cytokines upon re-stimulation, with lower levels or absence of Th2, Th17, and Th9 cytokines. Multivariate analyses indicated that a balanced memory T cell response with the production of Th1 and Th2 cytokines was associated with high levels of S1-specific binding and neutralizing antibodies mainly at 6 months after second vaccination. In conclusion, seroconversion is associated with the balanced production of cytokines by memory T cells. This emphasizes the importance of measuring multiple T cell cytokines to understand their influence on seroconversion and potentially gain more information about the protection induced by vaccine-induced memory T cells.
Collapse
Affiliation(s)
- Yvette den Hartog
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, The Netherlands
| | - S Reshwan K Malahe
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, The Netherlands
| | - Wim J R Rietdijk
- Department of Hospital Pharmacy, University Medical Center, Rotterdam, The Netherlands
| | - Marjolein Dieterich
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, The Netherlands
| | - Lennert Gommers
- Department of Viroscience, University Medical Center, Rotterdam, The Netherlands
| | - Daryl Geers
- Department of Viroscience, University Medical Center, Rotterdam, The Netherlands
| | - Susanne Bogers
- Department of Viroscience, University Medical Center, Rotterdam, The Netherlands
| | - Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Dimitri A Diavatopoulos
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - A Lianne Messchendorp
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Renate G van der Molen
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - Ester B M Remmerswaal
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Frederike J Bemelman
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ron T Gansevoort
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Jan-Stephan Sanders
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Marcia M L Kho
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, The Netherlands
| | - Marlies E J Reinders
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, The Netherlands
| | - Rory D de Vries
- Department of Viroscience, University Medical Center, Rotterdam, The Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
12
|
Westphal T, Mader M, Karsten H, Cords L, Knapp M, Schulte S, Hermanussen L, Peine S, Ditt V, Grifoni A, Addo MM, Huber S, Sette A, Lütgehetmann M, Pischke S, Kwok WW, Sidney J, Schulze zur Wiesch J. Evidence for broad cross-reactivity of the SARS-CoV-2 NSP12-directed CD4 + T-cell response with pre-primed responses directed against common cold coronaviruses. Front Immunol 2023; 14:1182504. [PMID: 37215095 PMCID: PMC10196118 DOI: 10.3389/fimmu.2023.1182504] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/06/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction The nonstructural protein 12 (NSP12) of the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) has a high sequence identity with common cold coronaviruses (CCC). Methods Here, we comprehensively assessed the breadth and specificity of the NSP12-specific T-cell response after in vitro T-cell expansion with 185 overlapping 15-mer peptides covering the entire SARS-CoV-2 NSP12 at single-peptide resolution in a cohort of 27 coronavirus disease 2019 (COVID-19) patients. Samples of nine uninfected seronegative individuals, as well as five pre-pandemic controls, were also examined to assess potential cross-reactivity with CCCs. Results Surprisingly, there was a comparable breadth of individual NSP12 peptide-specific CD4+ T-cell responses between COVID-19 patients (mean: 12.82 responses; range: 0-25) and seronegative controls including pre-pandemic samples (mean: 12.71 responses; range: 0-21). However, the NSP12-specific T-cell responses detected in acute COVID-19 patients were on average of a higher magnitude. The most frequently detected CD4+ T-cell peptide specificities in COVID-19 patients were aa236-250 (37%) and aa246-260 (44%), whereas the peptide specificities aa686-700 (50%) and aa741-755 (36%), were the most frequently detected in seronegative controls. In CCC-specific peptide-expanded T-cell cultures of seronegative individuals, the corresponding SARS-CoV-2 NSP12 peptide specificities also elicited responses in vitro. However, the NSP12 peptide-specific CD4+ T-cell response repertoire only partially overlapped in patients analyzed longitudinally before and after a SARS-CoV-2 infection. Discussion The results of the current study indicate the presence of pre-primed, cross-reactive CCC-specific T-cell responses targeting conserved regions of SARS-CoV-2, but they also underline the complexity of the analysis and the limited understanding of the role of the SARS-CoV-2 specific T-cell response and cross-reactivity with the CCCs.
Collapse
Affiliation(s)
- Tim Westphal
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Maria Mader
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hendrik Karsten
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leon Cords
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Knapp
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sophia Schulte
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lennart Hermanussen
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sven Peine
- Institute of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Ditt
- Institute of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Marylyn Martina Addo
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Institute of Infection Research and Vaccine Development, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Marc Lütgehetmann
- German Center for Infection Research Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sven Pischke
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - William W. Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Julian Schulze zur Wiesch
- Infectious Diseases Unit I, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research Deutsches Zentrum für Infektionsforschung (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
13
|
Vishnoi J, Sharma RK, Patel J, Sharma JC, Sharma KR, Mehta U. Severity and Outcome of Post-Vaccine COVID-19 among Healthcare Workers in a University Hospital in India. J Med Life 2023; 16:782-793. [PMID: 37520491 PMCID: PMC10375337 DOI: 10.25122/jml-2023-0017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/29/2023] [Indexed: 08/01/2023] Open
Abstract
Healthcare workers (HCWs) are at high risk of COVID-19 infection despite vaccination. Limited data exist on COVID-19 cases among vaccinated HCWs. This study aimed to describe the clinical characteristics and outcomes of RT PCR-confirmed COVID-19 cases in vaccinated HCWs, at a COVID clinic in a medical college hospital. This single-center, prospective cohort study included HCWs who received at least one dose of the COVID-19 vaccine and tested positive for COVID-19 within 6 months. Data on demographics, symptoms, work category, COVID-19 vaccination interval, and infection severity were collected. Of 2381 vaccinated HCWs, 105 tested positive and were categorized as mild, moderate, or severe cases. Among vaccinated HCWs, 4.41% had post-vaccine COVID-19 infections. All 105 cases received the first dose, and 79 received the second dose. Of the cases, 47.6% were partially vaccinated, and 53.3% were breakthrough cases. The mean age was 30.90±8.69 years, with 63.8% male and 36.2% female cases. Most cases (85.7%) acquired infection in the hospital, and 47.6% had direct contact with COVID-19 patients. Common symptoms included fatigue (85.7%), fever (82.9%), and cough (64.8%). Among cases, 93.3% were mild, 5.7% were moderate, and 0.9% were severe. Hospital admission and supplemental oxygen therapy were required for moderate and severe cases. No mortality was reported. Certain variables were associated with age, preventive measures, workplace type, symptoms, and comorbidities. Breakthrough infections can occur among fully vaccinated HCWs but with reduced severity and mortality. Monitoring and infection control measures remain crucial even in vaccinated individuals. This study provides insights into clinical presentations, oxygen therapy requirements, and outcomes of post-vaccine COVID-19 cases among HCWs. The data will inform strategies for booster doses to prevent COVID-19.
Collapse
Affiliation(s)
- Jagdish Vishnoi
- Department of Medicine, Pacific Medical College and Hospital, Pacific Medical University, Udaipur, Rajasthan, India
| | - Rajendra Kumar Sharma
- Department of Pediatrics, Pacific Medical College and Hospital, Pacific Medical University, Udaipur, Rajasthan, India
| | - Japan Patel
- Department of Medicine, Pacific Medical College and Hospital, Pacific Medical University, Udaipur, Rajasthan, India
| | - Jagdish Chandra Sharma
- Department of Medicine, Pacific Medical College and Hospital, Pacific Medical University, Udaipur, Rajasthan, India
| | - Kalu Ram Sharma
- Department of Medicine, Pacific Medical College and Hospital, Pacific Medical University, Udaipur, Rajasthan, India
| | - Urvansh Mehta
- Department of Medicine, Pacific Medical College and Hospital, Pacific Medical University, Udaipur, Rajasthan, India
| |
Collapse
|
14
|
Xu S, Li J, Wang H, Wang F, Yin Z, Wang Z. Real-world effectiveness and factors associated with effectiveness of inactivated SARS-CoV-2 vaccines: a systematic review and meta-regression analysis. BMC Med 2023; 21:160. [PMID: 37106390 PMCID: PMC10134725 DOI: 10.1186/s12916-023-02861-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND The two inactivated SARS-CoV-2 vaccines, CoronaVac and BBIBP-CorV, have been widely used to control the COVID-19 pandemic. The influence of multiple factors on inactivated vaccine effectiveness (VE) during long-term use and against variants is not well understood. METHODS We selected published or preprinted articles from PubMed, Embase, Scopus, Web of Science, medRxiv, BioRxiv, and the WHO COVID-19 database by 31 August 2022. We included observational studies that assessed the VE of completed primary series or homologous booster against SARS-CoV-2 infection or severe COVID-19. We used DerSimonian and Laird random-effects models to calculate pooled estimates and conducted multiple meta-regression with an information theoretic approach based on Akaike's Information Criterion to select the model and identify the factors associated with VE. RESULTS Fifty-one eligible studies with 151 estimates were included. For prevention of infection, VE associated with study region, variants, and time since vaccination; VE was significantly decreased against Omicron compared to Alpha (P = 0.021), primary series VE was 52.8% (95% CI, 43.3 to 60.7%) against Delta and 16.4% (95% CI, 9.5 to 22.8%) against Omicron, and booster dose VE was 65.2% (95% CI, 48.3 to 76.6%) against Delta and 20.3% (95% CI, 10.5 to 28.0%) against Omicron; primary VE decreased significantly after 180 days (P = 0.022). For the prevention of severe COVID-19, VE associated with vaccine doses, age, study region, variants, study design, and study population type; booster VE increased significantly (P = 0.001) compared to primary; though VE decreased significantly against Gamma (P = 0.034), Delta (P = 0.001), and Omicron (P = 0.001) compared to Alpha, primary and booster VEs were all above 60% against each variant. CONCLUSIONS Inactivated vaccine protection against SARS-CoV-2 infection was moderate, decreased significantly after 6 months following primary vaccination, and was restored by booster vaccination. VE against severe COVID-19 was greatest after boosting and did not decrease over time, sustained for over 6 months after the primary series, and more evidence is needed to assess the duration of booster VE. VE varied by variants, most notably against Omicron. It is necessary to ensure booster vaccination of everyone eligible for SARS-CoV-2 vaccines and continue monitoring virus evolution and VE. TRIAL REGISTRATION PROSPERO, CRD42022353272.
Collapse
Affiliation(s)
- Shiyao Xu
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China
| | - Jincheng Li
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China
| | - Hongyuan Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Fuzhen Wang
- Chinese Center for Disease Control and Prevention, National Immunization Programme, Beijing, China
| | - Zundong Yin
- Chinese Center for Disease Control and Prevention, National Immunization Programme, Beijing, China.
| | - Zhifeng Wang
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China.
| |
Collapse
|
15
|
Koopman G, Amacker M, Stegmann T, Verschoor EJ, Verstrepen BE, Bhoelan F, Bemelman D, Böszörményi KP, Fagrouch Z, Kiemenyi-Kayere G, Mortier D, Verel DE, Niphuis H, Acar RF, Kondova I, Kap YS, Bogers WMJM, Mooij P, Fleury S. A low dose of RBD and TLR7/8 agonist displayed on influenza virosome particles protects rhesus macaque against SARS-CoV-2 challenge. Sci Rep 2023; 13:5074. [PMID: 36977691 PMCID: PMC10044094 DOI: 10.1038/s41598-023-31818-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Influenza virosomes serve as antigen delivery vehicles and pre-existing immunity toward influenza improves the immune responses toward antigens. Here, vaccine efficacy was evaluated in non-human primates with a COVID-19 virosome-based vaccine containing a low dose of RBD protein (15 µg) and the adjuvant 3M-052 (1 µg), displayed together on virosomes. Vaccinated animals (n = 6) received two intramuscular administrations at week 0 and 4 and challenged with SARS-CoV-2 at week 8, together with unvaccinated control animals (n = 4). The vaccine was safe and well tolerated and serum RBD IgG antibodies were induced in all animals and in the nasal washes and bronchoalveolar lavages in the three youngest animals. All control animals became strongly sgRNA positive in BAL, while all vaccinated animals were protected, although the oldest vaccinated animal (V1) was transiently weakly positive. The three youngest animals had also no detectable sgRNA in nasal wash and throat. Cross-strain serum neutralizing antibodies toward Wuhan-like, Alpha, Beta, and Delta viruses were observed in animals with the highest serum titers. Pro-inflammatory cytokines IL-8, CXCL-10 and IL-6 were increased in BALs of infected control animals but not in vaccinated animals. Virosomes-RBD/3M-052 prevented severe SARS-CoV-2, as shown by a lower total lung inflammatory pathology score than control animals.
Collapse
Grants
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
- TRANSVAC2 2002-08-AVVAX-COVID-19, TRANSVAC2_TNA2002-08 European Commission
Collapse
Affiliation(s)
- Gerrit Koopman
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands.
| | - Mario Amacker
- Mymetics SA, 4 Route de La Corniche, 1066, Epalinges, Switzerland
- Department for BioMedical Research DBMR, Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, 3008, Bern, Switzerland
| | - Toon Stegmann
- Mymetics BV, JH Oortweg 21, 2333 CH, Leiden, The Netherlands
| | - Ernst J Verschoor
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Babs E Verstrepen
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Farien Bhoelan
- Mymetics BV, JH Oortweg 21, 2333 CH, Leiden, The Netherlands
| | - Denzel Bemelman
- Mymetics BV, JH Oortweg 21, 2333 CH, Leiden, The Netherlands
| | - Kinga P Böszörményi
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Zahra Fagrouch
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | | | - Daniella Mortier
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Dagmar E Verel
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Henk Niphuis
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Roja Fidel Acar
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Ivanela Kondova
- Animal Science Department, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Yolanda S Kap
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Willy M J M Bogers
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Petra Mooij
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Sylvain Fleury
- Mymetics SA, 4 Route de La Corniche, 1066, Epalinges, Switzerland.
| |
Collapse
|
16
|
Aaron Nathan Barksdale, Wood MG, Branecki CE, Zimmerman B, Lyden E, Nguyen TT, Hatfield A, Koepsell S, Langenfeld J, Zeger WG, Wadman MC. Incidence of unknown COVID-19 infection in a cohort of emergency physicians and advance practice providers. Am J Emerg Med 2023; 64:155-160. [PMID: 36563499 PMCID: PMC9749378 DOI: 10.1016/j.ajem.2022.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION In United States, health care workers have been immersed in the COVID-19 pandemic since February 2020. Since availability of COVID-19 vaccines, there is limited literature investigating the incidence of unknown COVID-19 infections in physicians and Advanced Practitioner Providers (APPs) working in emergency departments (EDs). The primary objective is to determine the incidence unknown COVID-19 infection within a cohort of emergency physicians (EPs) and APPs. METHODS Prospective observational study at a tertiary academic center with emergency medicine residency and 64,000 annual ED visits. EPs/APPs providing care to ED patients over the prior 12 months were eligible. Serum samples were collected between May 1 and June 30, 2022. Analysis utilized Luminex xMAP® SARS-CoV-2 Multi-Antigen IgG Assay for antibodies to Nucleocapsid, Receptor-binding domain, and Spike subunit 1. Mean Fluorescent Intensity (MFI) ≥ 700 was considered positive. Subjects completed 12 question survey assessing demographics and previously confirmed COVID-19 infection. Fisher's exact test evaluated associations of demographics and clinical characteristics with confirmed COVID-19 status. Analyses performed using SAS, Version 9.4. P < 0.05 considered statistically significant. RESULTS Sixty-nine of 81 eligible subjects (85.2%) participated, 58.0% were male, 97.1% white, with mean age of 37. Eighteen subjects had MFI ≥ 700 strongly suggestive of prior infection, with 17.7% unknown. No statistically significant difference between age, gender, race, children in home, or household member with previously COVID-19 infection. CONCLUSION Unknown previous COVID-19 infection was less then expected in this cohort of EPs/APPs, and no association with individual characteristics, previously infected household member, or children in the home.
Collapse
Affiliation(s)
- Aaron Nathan Barksdale
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America,Corresponding author at: 981150 Nebraska Medical Center, Omaha, NE 68198-1150, United States of America
| | - Macy G. Wood
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Chad E. Branecki
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Brooklin Zimmerman
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Elizabeth Lyden
- Research Design and Analysis, College of Public Health, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Thang T. Nguyen
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Andrew Hatfield
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Scott Koepsell
- Clinical Operations, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Jason Langenfeld
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Wesley G. Zeger
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Michael C. Wadman
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| |
Collapse
|
17
|
Dey RK, Imad HA, Aung PL, Faisham M, Moosa M, Hasna M, Afaa A, Ngamprasertchai T, Matsee W, Nguitragool W, Nakayama EE, Shioda T. Concurrent Infection with SARS-CoV-2 and Orientia tsutsugamushi during the COVID-19 Pandemic in the Maldives. Trop Med Infect Dis 2023; 8:82. [PMID: 36828498 PMCID: PMC9959419 DOI: 10.3390/tropicalmed8020082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
The COVID-19 pandemic was the worst public-health crisis in recent history. The impact of the pandemic in tropical regions was further complicated by other endemic tropical diseases, which can cause concurrent infections along with COVID-19. Here, we describe the clinical course of a patient with concurrent COVID-19 and scrub typhus infection. The patient's de-identified clinical data were retrieved retrospectively. The patient had progressive breathlessness at the time of presentation and was hospitalized for COVID-19. Respiratory examination revealed dyspnea, tachypnea, and coarse crepitations bilaterally over the entire lung field. Oxygenation was impaired, and a PaO2/FiO2 ratio of 229 suggested acute respiratory distress syndrome. Laboratory tests indicated leukocytosis, thrombocytopenia, ferritinemia, hypoalbuminemia, and transaminitis. Upon revaluation for persistent fever, physical examination revealed an eschar in the right antecubital fossa. Serology further confirmed scrub typhus, with IgM and IgG antibody positivity. A remarkable clinical recovery was achieved with doxycycline. The COVID-19 pandemic might have masked endemic tropical diseases. Clinicians working in endemic regions must always consider common tropical diseases that may present as a co-infection, as in our case. Travel and exposure history are critical guides for narrowing down a differential diagnosis. Early diagnosis and treatment can prevent complications.
Collapse
Affiliation(s)
- Rajib Kumar Dey
- Department of Medicine, Indira Gandhi Memorial Hospital, Malé 20002, Maldives
| | - Hisham Ahmed Imad
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Thai Travel Clinic, Hospital for Tropical Diseases, Bangkok 10400, Thailand
- Center for Infectious Disease Education and Research, Department of Viral Infections, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Pyae Linn Aung
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Mohamed Faisham
- Department of Medicine, Indira Gandhi Memorial Hospital, Malé 20002, Maldives
| | - Muaz Moosa
- Department of Medicine, Indira Gandhi Memorial Hospital, Malé 20002, Maldives
| | - Mariyam Hasna
- Department of Medicine, Indira Gandhi Memorial Hospital, Malé 20002, Maldives
| | - Aminath Afaa
- Department of Medicine, Indira Gandhi Memorial Hospital, Malé 20002, Maldives
| | - Thundon Ngamprasertchai
- Thai Travel Clinic, Hospital for Tropical Diseases, Bangkok 10400, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Wasin Matsee
- Thai Travel Clinic, Hospital for Tropical Diseases, Bangkok 10400, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Wang Nguitragool
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Emi E. Nakayama
- Center for Infectious Disease Education and Research, Department of Viral Infections, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Tatsuo Shioda
- Center for Infectious Disease Education and Research, Department of Viral Infections, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
18
|
Schnizer C, Andreas N, Vivas W, Kamradt T, Baier M, Kiehntopf M, Glöckner S, Scherag A, Löffler B, Kolanos S, Guerra J, Pletz MW, Weis S. Persistent humoral and CD4 + T H cell immunity after mild SARS-COV-2 infection-The CoNAN long-term study. Front Immunol 2023; 13:1095129. [PMID: 36713390 PMCID: PMC9880277 DOI: 10.3389/fimmu.2022.1095129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023] Open
Abstract
Understanding persistent cellular and humoral immune responses to SARS-CoV-2 will be of major importance to terminate the ongoing pandemic. Here, we assessed long-term immunity in individuals with mild COVID-19 up to 1 year after a localized SARS-CoV-2 outbreak. CoNAN was a longitudinal population-based cohort study performed 1.5 months, 6 months, and 12 months after a SARS-CoV-2 outbreak in a rural German community. We performed a time series of five different IgG immunoassays assessing SARS-CoV-2 antibody responses on serum samples from individuals that had been tested positive after a SARS-CoV-2 outbreak and in control individuals who had a negative PCR result. These analyses were complemented with the determination of spike-antigen specific TH cell responses in the same individuals. All infected participants were presented as asymptomatic or mild cases. Participants initially tested positive for SARS-CoV-2 infection either with PCR, antibody testing, or both had a rapid initial decline in the serum antibody levels in all serological tests but showed a persisting TH cell immunity as assessed by the detection of SARS-CoV-2 specificity of TH cells for up to 1 year after infection. Our data support the notion of a persistent T-cell immunity in mild and asymptomatic cases of SARS-CoV-2 up to 1 year after infection. We show that antibody titers decline over 1 year, but considering several test results, complete seroreversion is rare. Trial registration German Clinical Trials Register DRKS00022416.
Collapse
Affiliation(s)
- Clara Schnizer
- Institute for Infectious Diseases and Infection Control, Jena University Hospital- Friedrich Schiller University, Jena, Germany
| | - Nico Andreas
- Institute of Immunology, Jena University Hospital - Friedrich Schiller University, Jena, Germany
| | - Wolfgang Vivas
- Institute for Infectious Diseases and Infection Control, Jena University Hospital- Friedrich Schiller University, Jena, Germany,Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena, Germany,Department of Anesthesiology and Intensive Care, Jena University Hospital- Friedrich Schiller University, Jena, Germany
| | - Thomas Kamradt
- Institute of Immunology, Jena University Hospital - Friedrich Schiller University, Jena, Germany
| | - Michael Baier
- Institute of Medical Microbiology, Jena University Hospital- Friedrich Schiller University, Jena, Germany
| | - Michael Kiehntopf
- Institute of Clinical Chemistry and Laboratory Diagnostics and Integrated Biobank Jena (IBBJ), Jena University Hospital- Friedrich Schiller University, Jena, Germany
| | - Stefan Glöckner
- Institute of Medical Microbiology, Jena University Hospital- Friedrich Schiller University, Jena, Germany
| | - André Scherag
- Institute of Medical Statistics, Computer and Data Sciences, Jena University Hospital- Friedrich Schiller University, Jena, Germany
| | - Bettina Löffler
- Institute of Medical Microbiology, Jena University Hospital- Friedrich Schiller University, Jena, Germany
| | - Steffi Kolanos
- Institute for Infectious Diseases and Infection Control, Jena University Hospital- Friedrich Schiller University, Jena, Germany
| | - Joel Guerra
- Institute for Infectious Diseases and Infection Control, Jena University Hospital- Friedrich Schiller University, Jena, Germany,Department of Anesthesiology and Intensive Care, Jena University Hospital- Friedrich Schiller University, Jena, Germany
| | - Mathias W. Pletz
- Institute for Infectious Diseases and Infection Control, Jena University Hospital- Friedrich Schiller University, Jena, Germany,Center for Sepsis Control and Care, Jena University Hospital- Friedrich Schiller University, Jena, Germany,*Correspondence: Sebastian Weis, ; Mathias W. Pletz,
| | - Sebastian Weis
- Institute for Infectious Diseases and Infection Control, Jena University Hospital- Friedrich Schiller University, Jena, Germany,Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena, Germany,Department of Anesthesiology and Intensive Care, Jena University Hospital- Friedrich Schiller University, Jena, Germany,*Correspondence: Sebastian Weis, ; Mathias W. Pletz,
| |
Collapse
|
19
|
Sikkema RS, de Bruin E, Ramakers C, Bentvelsen R, Li W, Bosch BJ, Westerhuis B, Haagmans B, Koopmans MPG, Fraaij PLA. Reduced Seasonal Coronavirus Antibody Responses in Children Following COVID-19 Mitigation Measures, The Netherlands. Viruses 2023; 15:212. [PMID: 36680252 PMCID: PMC9862716 DOI: 10.3390/v15010212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
SARS-CoV-2 prevention and control measures did not only impact SARS-CoV-2 circulation, but also the timing and prevalence of other seasonal respiratory viruses. Especially in children, information on exposure and infections to seasonal coronaviruses as well as SARS-CoV-2 in the first year of the pandemic is largely lacking. Therefore, we set up a one-year serological survey in a large tertiary hospital in the Netherlands. We show that seasonal coronavirus seroprevalence significantly decreased in 2021 in children less than one year, most likely due to COVID-19 control measures. The SARS-CoV-2 seroprevalence in children and adolescents increased from 0.4% to 11.3%, the highest in adolescents. This implies higher exposure rates in adolescents as compared to the general population (>18 years old). It is clear that there have been significant changes in the circulation and subsequent immunity against most respiratory pathogens as a result of the mitigation measures. The implications on shorter as well as longer term are still largely unknown, but the impact of the SARS-CoV-2 pandemic and subsequent control measures will continue to affect the dynamics of other pathogens.
Collapse
Affiliation(s)
| | - Erwin de Bruin
- Viroscience, Erasmus MC, 3015 Rotterdam, The Netherlands
| | | | - Robbert Bentvelsen
- Microvida Laboratory for Microbiology, Amphia Hospital, 4818 Breda, The Netherlands
- Department of Medical Microbiology, Leiden University Medical Center, 2333 Leiden, The Netherlands
| | - Wentao Li
- Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 Utrecht, The Netherlands
| | - Berend-Jan Bosch
- Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 Utrecht, The Netherlands
| | | | - Bart Haagmans
- Viroscience, Erasmus MC, 3015 Rotterdam, The Netherlands
| | | | - Pieter L. A. Fraaij
- Viroscience, Erasmus MC, 3015 Rotterdam, The Netherlands
- Pediatrics, Erasmus MC-Sophia Children’s Hospital, 3015 Rotterdam, The Netherlands
| |
Collapse
|
20
|
Taffertshofer K, Walter M, Mackeben P, Kraemer J, Potapov S, Jochum S. Design and performance characteristics of the Elecsys anti-SARS-CoV-2 S assay. Front Immunol 2022; 13:1002576. [PMID: 36532081 PMCID: PMC9756759 DOI: 10.3389/fimmu.2022.1002576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Background Automated, high throughput assays are required to quantify the immune response after infection with or vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This study on the Roche Elecsys® Anti-SARS-CoV-2 S (ACOV2S) assay provides insights on the assay design and performance. Methods The ACOV2S assay quantifies antibodies to the receptor-binding domain of the SARS-CoV-2 spike protein. The assigned units and the underlying standardization were compared to the international reference standard in BAU/mL. Assay specificity was assessed in samples (n=5981) collected prior to the COVID-19 pandemic and in samples from patients with non-COVID-19 respiratory infections (n=697) or other infectious diseases (n=771). Sensitivity was measured in 1313 samples from patients with mild COVID-19 and 297 samples from patients hospitalized with COVID-19. Comparison of results was performed to a comparator semi-quantitative anti-S1 assay of indirect detection format as well as a commercially available and an in-house version of a surrogate neutralization assay (ACE2-RBD). Results The originally assigned units for the ACOV2S assay were shown to be congruent to the units of the First International WHO Standard for anti-SARS-CoV-2 immunoglobulins. Overall specificity was 99.98% with no geographical differences noted and no loss of specificity in samples containing potentially cross-reacting antibodies. High sensitivity was observed, with 98.8% of samples reported to be reactive >14 days after infection and sustained detection of antibodies over time. For all samples, ACOV2S titers and neutralization capacities developed with comparable dynamics. Robust standardization and assay setup enable excellent reproducibility of results, independent of lot or analyzer used. Conclusion The results from this study confirmed that ACOV2S is a highly sensitive and specific assay and correlates well with surrogate neutralization assays. The units established for ACOV2S are also interchangeable with the units of the First International WHO Standard for anti-SARS-CoV-2 immunoglobulins. Worldwide availability of the assay and analyzers render ACOV2S a highly practical tool for population-wide assessment and monitoring of the humoral response to SARS-CoV-2 infection or vaccination.
Collapse
Affiliation(s)
- Karin Taffertshofer
- Research and Development Immunoassays, Roche Diagnostics GmbH, Penzberg, Germany
| | - Mirko Walter
- Research and Development Immunoassays, Roche Diagnostics GmbH, Penzberg, Germany
| | - Peter Mackeben
- Research and Development Immunoassays, Roche Diagnostics GmbH, Penzberg, Germany
| | - Julia Kraemer
- Research and Development Immunoassays, Roche Diagnostics GmbH, Penzberg, Germany
| | - Sergej Potapov
- Biostatistics & Data Science, Roche Diagnostics GmbH, Penzberg, Germany
| | - Simon Jochum
- Research and Development Immunoassays, Roche Diagnostics GmbH, Penzberg, Germany,*Correspondence: Simon Jochum,
| |
Collapse
|
21
|
Contributions of vibrational spectroscopy to virology: A review. CLINICAL SPECTROSCOPY 2022; 4:100022. [PMCID: PMC9093054 DOI: 10.1016/j.clispe.2022.100022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/30/2022] [Accepted: 05/04/2022] [Indexed: 06/17/2023]
Abstract
Vibrational spectroscopic techniques, both infrared absorption and Raman scattering, are high precision, label free analytical techniques which have found applications in fields as diverse as analytical chemistry, pharmacology, forensics and archeometrics and, in recent times, have attracted increasing attention for biomedical applications. As analytical techniques, they have been applied to the characterisation of viruses as early as the 1970 s, and, in the context of the coronavirus disease 2019 (COVID-19) pandemic, have been explored in response to the World Health Organisation as novel methodologies to aid in the global efforts to implement and improve rapid screening of viral infection. This review considers the history of the application of vibrational spectroscopic techniques to the characterisation of the morphology and chemical compositions of viruses, their attachment to, uptake by and replication in cells, and their potential for the detection of viruses in population screening, and in infection response monitoring applications. Particular consideration is devoted to recent efforts in the detection of severe acute respiratory syndrome coronavirus 2, and monitoring COVID-19.
Collapse
|
22
|
Thompson EJ, Stafford J, Moltrecht B, Huggins CF, Kwong ASF, Shaw RJ, Zaninotto P, Patel K, Silverwood RJ, McElroy E, Pierce M, Green MJ, Bowyer RCE, Maddock J, Tilling K, Katikireddi SV, Ploubidis GB, Porteous DJ, Timpson N, Chaturvedi N, Steves CJ, Patalay P. Psychological distress, depression, anxiety, and life satisfaction following COVID-19 infection: evidence from 11 UK longitudinal population studies. Lancet Psychiatry 2022; 9:894-906. [PMID: 36244359 PMCID: PMC9560745 DOI: 10.1016/s2215-0366(22)00307-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/14/2022] [Accepted: 08/09/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Evidence on associations between COVID-19 illness and mental health is mixed. We aimed to examine whether COVID-19 is associated with deterioration in mental health while considering pre-pandemic mental health, time since infection, subgroup differences, and confirmation of infection via self-reported test and serology data. METHODS We obtained data from 11 UK longitudinal studies with repeated measures of mental health (psychological distress, depression, anxiety, and life satisfaction; mental health scales were standardised within each study across time) and COVID-19 status between April, 2020, and April, 2021. We included participants with information available on at least one mental health outcome measure and self-reported COVID-19 status (suspected or test-confirmed) during the pandemic, and a subset with serology-confirmed COVID-19. Furthermore, only participants who had available data on a minimum set of covariates, including age, sex, and pre-pandemic mental health were included. We investigated associations between having ever had COVID-19 and mental health outcomes using generalised estimating equations. We examined whether associations varied by age, sex, ethnicity, education, and pre-pandemic mental health, whether the strength of the association varied according to time since infection, and whether associations differed between self-reported versus confirmed (by test or serology) infection. FINDINGS Between 21 Dec, 2021, and July 11, 2022, we analysed data from 54 442 participants (ranging from a minimum age of 16 years in one study to a maximum category of 90 years and older in another; including 33 200 [61·0%] women and 21 242 [39·0%] men) from 11 longitudinal UK studies. Of 40 819 participants with available ethnicity data, 36 802 (90·2%) were White. Pooled estimates of standardised differences in outcomes suggested associations between COVID-19 and subsequent psychological distress (0·10 [95% CI 0·06 to 0·13], I2=42·8%), depression (0·08 [0·05 to 0·10], I2=20·8%), anxiety (0·08 [0·05 to 0·10], I2=0·0%), and lower life satisfaction (-0·06 [-0·08 to -0·04], I2=29·2%). We found no evidence of interactions between COVID-19 and sex, education, ethnicity, or pre-pandemic mental health. Associations did not vary substantially between time since infection of less than 4 weeks, 4-12 weeks, and more than 12 weeks, and were present in all age groups, with some evidence of stronger effects in those aged 50 years and older. Participants who self-reported COVID-19 but had negative serology had worse mental health outcomes for all measures than those without COVID-19 based on serology and self-report. Participants who had positive serology but did not self-report COVID-19 did not show association with mental health outcomes. INTERPRETATION Self-reporting COVID-19 was longitudinally associated with deterioration in mental health and life satisfaction. Our findings emphasise the need for greater post-infection mental health service provision, given the substantial prevalence of COVID-19 in the UK and worldwide. FUNDING UK Medical Research Council and UK National Institute for Health and Care Research.
Collapse
Affiliation(s)
- Ellen J Thompson
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, UK
| | - Jean Stafford
- MRC Unit for Lifelong Health and Ageing, University College London, London, UK.
| | - Bettina Moltrecht
- Centre for Longitudinal Studies, University College London, London, UK
| | - Charlotte F Huggins
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
| | - Alex S F Kwong
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Richard J Shaw
- MRC/CSO Social & Public Health Sciences Unit, School of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Paola Zaninotto
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Kishan Patel
- MRC Unit for Lifelong Health and Ageing, University College London, London, UK
| | | | - Eoin McElroy
- School of Psychology, Ulster University, Ulster, UK
| | - Matthias Pierce
- Division of Psychology and Mental Health, The University of Manchester, Manchester, UK
| | - Michael J Green
- MRC/CSO Social & Public Health Sciences Unit, School of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Ruth C E Bowyer
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, UK
| | - Jane Maddock
- MRC Unit for Lifelong Health and Ageing, University College London, London, UK
| | - Kate Tilling
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - S Vittal Katikireddi
- MRC/CSO Social & Public Health Sciences Unit, School of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | | | - David J Porteous
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
| | - Nic Timpson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Nish Chaturvedi
- MRC Unit for Lifelong Health and Ageing, University College London, London, UK
| | - Claire J Steves
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, UK
| | - Praveetha Patalay
- MRC Unit for Lifelong Health and Ageing, University College London, London, UK; Centre for Longitudinal Studies, University College London, London, UK
| |
Collapse
|
23
|
Yaugel-Novoa M, Bourlet T, Paul S. Role of the humoral immune response during COVID-19: guilty or not guilty? Mucosal Immunol 2022; 15:1170-1180. [PMID: 36195658 PMCID: PMC9530436 DOI: 10.1038/s41385-022-00569-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/07/2022] [Accepted: 09/19/2022] [Indexed: 02/04/2023]
Abstract
Systemic and mucosal humoral immune responses are crucial to fight respiratory viral infections in the current pandemic of COVID-19 caused by the SARS-CoV-2 virus. During SARS-CoV-2 infection, the dynamics of systemic and mucosal antibody infections are affected by patient characteristics, such as age, sex, disease severity, or prior immunity to other human coronaviruses. Patients suffering from severe disease develop higher levels of anti-SARS-CoV-2 antibodies in serum and mucosal tissues than those with mild disease, and these antibodies are detectable for up to a year after symptom onset. In hospitalized patients, the aberrant glycosylation of anti-SARS-CoV-2 antibodies enhances inflammation-associated antibody Fc-dependent effector functions, thereby contributing to COVID-19 pathophysiology. Current vaccines elicit robust humoral immune responses, principally in the blood. However, they are less effective against new viral variants, such as Delta and Omicron. This review provides an overview of current knowledge about the humoral immune response to SARS-CoV-2, with a particular focus on the protective and pathological role of humoral immunity in COVID-19 severity. We also discuss the humoral immune response elicited by COVID-19 vaccination and protection against emerging viral variants.
Collapse
Affiliation(s)
- Melyssa Yaugel-Novoa
- CIRI—Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Université Claude Bernard Lyon 1, Lyon, France
| | - Thomas Bourlet
- CIRI—Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Université Claude Bernard Lyon 1, Lyon, France
| | - Stéphane Paul
- CIRI—Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Université Claude Bernard Lyon 1, Lyon, France,CIC Inserm 1408 Vaccinology, Saint-Etienne, France
| |
Collapse
|
24
|
Mallory RM, Formica N, Pfeiffer S, Wilkinson B, Marcheschi A, Albert G, McFall H, Robinson M, Plested JS, Zhu M, Cloney-Clark S, Zhou B, Chau G, Robertson A, Maciejewski S, Hammond HL, Baracco L, Logue J, Frieman MB, Smith G, Patel N, Glenn GM. Safety and immunogenicity following a homologous booster dose of a SARS-CoV-2 recombinant spike protein vaccine (NVX-CoV2373): a secondary analysis of a randomised, placebo-controlled, phase 2 trial. THE LANCET. INFECTIOUS DISEASES 2022; 22:1565-1576. [PMID: 35963274 PMCID: PMC9365313 DOI: 10.1016/s1473-3099(22)00420-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/01/2022] [Accepted: 06/09/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Emerging SARS-CoV-2 variants and evidence of waning vaccine efficacy present substantial obstacles towards controlling the COVID-19 pandemic. Booster doses of SARS-CoV-2 vaccines might address these concerns by amplifying and broadening the immune responses seen with initial vaccination regimens. We aimed to assess the immunogenicity and safety of a homologous booster dose of a SARS-CoV-2 recombinant spike protein vaccine (NVX-CoV2373). METHODS This secondary analysis of a phase 2, randomised study assessed a single booster dose of a SARS-CoV-2 recombinant spike protein vaccine with Matrix-M adjuvant (NVX-CoV2373) in healthy adults aged 18-84 years, recruited from 17 clinical centres in the USA and Australia. Eligible participants had a BMI of 17-35 kg/m2 and, for women, were heterosexually inactive or using contraception. Participants who had a history of SARS-CoV or SARS-CoV-2, confirmed diagnosis of COVID-19, serious chronic medical conditions, or were pregnant or breastfeeding were excluded. Approximately 6 months following their primary two-dose vaccination series (administered day 0 and day 21), participants who received placebo for their primary vaccination series received a placebo booster (group A) and participants who received NVX-CoV2373 for their primary vaccination series (group B) were randomly assigned (1:1) again, via centralised interactive response technology system, to receive either placebo (group B1) or a single booster dose of NVX-CoV2373 (5 μg SARS-CoV-2 rS with 50 μg Matrix-M adjuvant; group B2) via intramuscular injection; randomisation was stratified by age and study site. Vaccinations were administered by designated site personnel who were masked to treatment assignment, and participants and other site staff were also masked. Administration personnel also assessed the outcome. The primary endpoints are safety (unsolicited adverse events) and reactogenicity (solicited local and systemic) events and immunogenicity (serum IgG antibody concentrations for the SARS-CoV-2 rS protein antigen) assessed 14 days after the primary vaccination series (day 35) and 28 days following booster (day 217). Safety was analysed in all participants in groups A, B1, and B2, according to the treatment received; immunogenicity was analysed in the per-protocol population (ie, participants in groups A, B1, and B2) who received all assigned doses and who did not test SARS-CoV-2-positive or received an authorised vaccine, analysed according to treatment assignment). This trial is registered with ClinicalTrials.gov, NCT04368988. FINDINGS 1610 participants were screened from Aug 24, 2020, to Sept 25, 2020. 1282 participants were enrolled, of whom 173 were assigned again to placebo (group A), 106 were re-randomised to NVX-CoV2373-placebo (group B1), and 104 were re-randomised to NVX-CoV2373-NVX-CoV2373 (group B2); after accounting for exclusions and incorrect administration, 172 participants in group A, 102 in group B1, and 105 in group B2 were analysed for safety. Following the active booster, the proportion of participants with available data reporting local (80 [82%] of 97 participants had any adverse event; 13 [13%] had a grade ≥3 event) and systemic (75 [77%] of 98 participants had any adverse event; 15 [15%] had a grade ≥3 event) reactions was higher than after primary vaccination (175 [70%] of 250 participants had any local adverse event, 13 [5%] had a grade ≥3 event; 132 [53%] of 250 had any systemic adverse event, 14 [6%] had a grade ≥3 event). Local and systemic events were transient in nature (median duration 1·0-2·5 days). In the per-protocol immunogenicity population at day 217 (167 participants in group A, 101 participants in group B1, 101 participants in group B2), IgG geometric mean titres (GMT) had increased by 4·7-fold and MN50 GMT by 4·1-fold for the ancestral SARS-CoV-2 strain compared with the day 35 titres. INTERPRETATION Administration of a booster dose of NVX-CoV2373 resulted in an incremental increase in reactogenicity. For both the prototype strain and all variants evaluated, immune responses following the booster were similar to or higher than those associated with high levels of efficacy in phase 3 studies of the vaccine. These data support the use of NVX-CoV2373 in booster programmes. FUNDING Novavax and the Coalition for Epidemic Preparedness Innovations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Holly L Hammond
- University of Maryland School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Lauren Baracco
- University of Maryland School of Medicine, University of Maryland, Baltimore, MD, USA
| | - James Logue
- University of Maryland School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Matthew B Frieman
- University of Maryland School of Medicine, University of Maryland, Baltimore, MD, USA
| | | | | | | |
Collapse
|
25
|
Importance of the COVID-19 Vaccine Booster Dose in Protection and Immunity. Vaccines (Basel) 2022; 10:vaccines10101708. [PMID: 36298573 PMCID: PMC9610198 DOI: 10.3390/vaccines10101708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022] Open
Abstract
Background: There is debate on the necessity of booster doses of COVID-19 vaccination, especially in countries with limited resources. Methods: This cross-sectional study was conducted in a referral laboratory in Tehran, Iran. The level of COVID-19 antibodies was measured and compared between individuals regarding the number of COVID-19 vaccine shots. Results: In this study, 176 individuals with a mean age of 36.3 (±11.7) years participated. A total of 112 individuals received two doses of the COVID-19 vaccine, and 64 individuals received three doses. Level of all antibodies was higher in those who received three doses than in those who received two doses of the COVID-19 vaccine. Considering the SARS-CoV-2 Spike IgG, the difference was not statistically significant but for the SARS-CoV-2 RBD IgG and SARS-CoV-2 NAB the difference was statistically significant. Regarding to the background variables, receiving influenza vaccine in the past year, history of autoimmune diseases and past medical history of chicken pox showed a significant association with the number of vaccine doses received. Their effects on the outcome variables assessed with multivariate logistic regression analysis. Conclusion: The results of our study show that a booster dose of the COVID-19 vaccine enhances the antibody response.
Collapse
|
26
|
How Will the COVID-19 Pandemic End? JOURNAL OF MEDICAL MICROBIOLOGY AND INFECTIOUS DISEASES 2022. [DOI: 10.52547/jommid.10.3.146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
27
|
Salum SS, Sheikh MA, Hebestreit A, Kelm S. Anti SARS-CoV-2 seroprevalence in Zanzibar in 2021 before the Omicron wave. IJID REGIONS 2022; 4:120-122. [PMID: 35814620 PMCID: PMC9251954 DOI: 10.1016/j.ijregi.2022.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/22/2022] [Accepted: 06/26/2022] [Indexed: 12/17/2022]
Abstract
Objectives For Tanzania, including Zanzibar, the development of the COVID-19 pandemic has remained unclear since the reporting of cases was suspended during 2020/21. Our study was the first to analyze data on COVID-19 seroprevalence in the Zanzibari population before the Omicron variant wave began in late 2021. Design During August through October 2021, representative cross-sectional data were collected from randomly selected households in 120 wards of the two main islands, Unguja and Pemba. Participants voluntarily provided blood samples to test their sera for antibodies against SARS-CoV-2 using a semiquantitative enzyme-linked immunosorbent assay (ELISA). Results 58.9% of the 2051 sera analysed were positive, without significant differences between Unguja and Pemba or between rural and urban areas. The results were in agreement with observations from other sub-Saharan African countries. Conclusions The antibody levels observed were most likely due to previous infections with SARS-CoV-2, since vaccination was generally not available before the survey. Therefore, this study offers the first insights into how many Zanzibari had COVID-19 before the Omicron variant emerged. Furthermore, it provides an appropriate basis for a follow-up survey addressing how this seroprevalence has influenced susceptibility to the Omicron variants, given the use of harmonized methodologies.
Collapse
Affiliation(s)
- Salum Seif Salum
- Department of Pathology, School of Health and Medical Sciences, State University of Zanzibar, Zanzibar, Tanzania
| | - Mohammed Ali Sheikh
- Environmental Analytical Chemistry and Eco‐toxicology Lab, State University of Zanzibar, Zanzibar, Tanzania
| | - Antje Hebestreit
- Leibniz Institute for Prevention Research and Epidemiology — BIPS, Bremen, Germany
| | - Sørge Kelm
- Centre for Biomolecular Interactions Bremen, Faculty for Biology and Chemistry, University Bremen, Bremen, Germany
| |
Collapse
|
28
|
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
|
29
|
Asymmetric Mach–Zehnder Interferometric Biosensing for Quantitative and Sensitive Multiplex Detection of Anti-SARS-CoV-2 Antibodies in Human Plasma. BIOSENSORS 2022; 12:bios12080553. [PMID: 35892450 PMCID: PMC9394312 DOI: 10.3390/bios12080553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 12/11/2022]
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic has once more emphasized the urgent need for accurate and fast point-of-care (POC) diagnostics for outbreak control and prevention. The main challenge in the development of POC in vitro diagnostics (IVD) is to combine a short time to result with a high sensitivity, and to keep the testing cost-effective. In this respect, sensors based on photonic integrated circuits (PICs) may offer advantages as they have features such as a high analytical sensitivity, capability for multiplexing, ease of miniaturization, and the potential for high-volume manufacturing. One special type of PIC sensor is the asymmetric Mach–Zehnder Interferometer (aMZI), which is characterized by a high and tunable analytical sensitivity. The current work describes the application of an aMZI-based biosensor platform for sensitive and multiplex detection of anti-SARS-CoV-2 antibodies in human plasma samples using the spike protein (SP), the receptor-binding domain (RBD), and the nucleocapsid protein (NP) as target antigens. The results are in good agreement with several CE-IVD marked reference methods and demonstrate the potential of the aMZI biosensor technology for further development into a photonic IVD platform.
Collapse
|
30
|
Sparrow E, Hasso-Agopsowicz M, Kaslow DC, Singh K, Rao R, Chibi M, Makubalo LE, Reeder JC, Kang G, Karron RA, Cravioto A, Lanata CF, Friede M, Abela-Ridder B, Solomon AW, Dagne DA, Giersing B. Leveraging mRNA Platform Technology to Accelerate Development of Vaccines for Some Emerging and Neglected Tropical Diseases Through Local Vaccine Production. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.844039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The mRNA vaccine technology platform may enable rapid response to some emerging infectious diseases (EIDs), as demonstrated through the COVID-19 pandemic. Beyond the role it could play in future EID response, mRNA technology also could have an important role in accelerating the development of, and access to, vaccines for some neglected tropical diseases (NTDs), which occur mainly in impoverished regions of the world. Despite their significant disease burden, few vaccines against NTDs have been developed, in part because of the uncertain market and return on investment. In addition, the probability of technical and regulatory success is considered to be low for developing vaccines against multicellular parasites, or organisms that have sophisticated mechanisms for evading immunological surveillance, such as many of the NTD pathogens. The global 2021-2030 road map for neglected tropical diseases sets ambitious targets for the eradication, elimination, and control of NTDs. For some, effective interventions exist but are underutilized. For others, vaccines need to be developed or their use expanded to meet global targets on control and elimination. This article discusses the application of the mRNA technology platform to the development of vaccines for NTDs as well as EIDs, highlights the challenges in bringing these products to the market, and indicates potential areas which could be explored, including leveraging investment for vaccines with a more profitable market potential and enabling local manufacturing in regions where NTDs are endemic. Such regional production could include collaborations with the mRNA vaccine technology transfer hubs that are being established with the support of WHO and COVAX partners.
Collapse
|
31
|
Cruz-Machado AR, Barreira SC, Bandeira M, Veldhoen M, Gomes A, Serrano M, Duarte C, Rato M, Miguel Fernandes B, Garcia S, Pinheiro F, Bernardes M, Madeira N, Miguel C, Torres R, Bento Silva A, Pestana J, Almeida D, Mazeda C, Cunha Santos F, Pinto P, Sousa M, Parente H, Sequeira G, Santos MJ, Fonseca JE, Romão VC. Risk Factors for Infection, Predictors of Severe Disease, and Antibody Response to COVID-19 in Patients With Inflammatory Rheumatic Diseases in Portugal-A Multicenter, Nationwide Study. Front Med (Lausanne) 2022; 9:901817. [PMID: 35770002 PMCID: PMC9234392 DOI: 10.3389/fmed.2022.901817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/16/2022] [Indexed: 11/30/2022] Open
Abstract
Objective To identify risk factors for SARS-CoV-2 infection and for severe/critical COVID-19, and to assess the humoral response after COVID-19 in these patients. Methods Nationwide study of adult patients with inflammatory RMDs prospectively followed in the Rheumatic Diseases Portuguese Register-Reuma.pt-during the first 6 months of the pandemic. We compared patients with COVID-19 with those who did not develop the disease and patients with mild/moderate disease with those exhibiting severe/critical COVID-19. IgG antibodies against SARS-CoV-2 were measured ≥3 months after infection and results were compared with matched controls. Results 162 cases of COVID-19 were registered in a total of 6,363 appointments. Patients treated with TNF inhibitors (TNFi; OR = 0.160, 95% CI 0.099-0.260, P < 0.001) and tocilizumab (OR 0.147, 95% CI 0.053-0.408, P < 0.001) had reduced odds of infection. Further, TNFi tended to be protective of severe and critical disease. Older age, major comorbidities, and rituximab were associated with an increased risk of infection and worse prognosis. Most patients with inflammatory RMDs (86.2%) developed a robust antibody response. Seroconversion was associated with symptomatic disease (OR 13.46, 95% CI 2.21-81.85, P = 0.005) and tended to be blunted by TNFi (OR 0.17, 95% CI 0.03-1.05; P = 0.057). Conclusions TNFi and tocilizumab reduced the risk of infection by SARS-CoV-2. Treatment with TNFi also tended to reduce rates of severe disease and seroconversion. Older age, general comorbidities and rituximab were associated with increased risk for infection and worse prognosis, in line with previous reports. Most patients with RMDs developed a proper antibody response after COVID-19, particularly if they had symptomatic disease.
Collapse
Affiliation(s)
- Ana Rita Cruz-Machado
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon Academic Medical Center and European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ERN-ReCONNET), Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sofia C. Barreira
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon Academic Medical Center and European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ERN-ReCONNET), Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Matilde Bandeira
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon Academic Medical Center and European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ERN-ReCONNET), Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Marc Veldhoen
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Andreia Gomes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Marta Serrano
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Catarina Duarte
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon Academic Medical Center and European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ERN-ReCONNET), Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Rato
- Rheumatology Department, Centro Hospitalar Universitário de São João EPE, Porto, Portugal
| | - Bruno Miguel Fernandes
- Rheumatology Department, Centro Hospitalar Universitário de São João EPE, Porto, Portugal
| | - Salomé Garcia
- Rheumatology Department, Centro Hospitalar Universitário de São João EPE, Porto, Portugal
| | - Filipe Pinheiro
- Rheumatology Department, Centro Hospitalar Universitário de São João EPE, Porto, Portugal
| | - Miguel Bernardes
- Rheumatology Department, Centro Hospitalar Universitário de São João EPE, Porto, Portugal
| | - Nathalie Madeira
- Rheumatology Department, Instituto Português de Reumatologia, Lisbon, Portugal
| | - Cláudia Miguel
- Rheumatology Department, Instituto Português de Reumatologia, Lisbon, Portugal
| | - Rita Torres
- Rheumatology Department, Hospital de Egas Moniz, Centro Hospitalar Lisboa Ocidental, Lisbon, Portugal
| | - Ana Bento Silva
- Rheumatology Department, Hospital de Egas Moniz, Centro Hospitalar Lisboa Ocidental, Lisbon, Portugal
| | - Jorge Pestana
- Rheumatology Department, Hospital Garcia de Orta, Almada, Portugal
| | - Diogo Almeida
- Rheumatology Department, Hospital de Braga, Braga, Portugal
| | - Carolina Mazeda
- Rheumatology Department, Centro Hospitalar do Baixo Vouga and iBiMED, Institute for Biomedicine, University of Aveiro, Aveiro, Portugal
| | | | - Patrícia Pinto
- Rheumatology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Marlene Sousa
- Rheumatology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Hugo Parente
- Rheumatology Department, Unidade Local de Saúde do Alto Minho, Ponte de Lima, Portugal
| | - Graça Sequeira
- Rheumatology Department, Centro Hospitalar Universitário do Algarve, Faro, Portugal
| | | | - João Eurico Fonseca
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon Academic Medical Center and European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ERN-ReCONNET), Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Vasco C. Romão
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon Academic Medical Center and European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ERN-ReCONNET), Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
32
|
Cheng H, Peng Z, Si S, Alifu X, Zhou H, Chi P, Zhuang Y, Mo M, Yu Y. Immunogenicity and Safety of Homologous and Heterologous Prime-Boost Immunization with COVID-19 Vaccine: Systematic Review and Meta-Analysis. Vaccines (Basel) 2022; 10:798. [PMID: 35632554 PMCID: PMC9142990 DOI: 10.3390/vaccines10050798] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/13/2022] Open
Abstract
A prime-boost strategy of COVID-19 vaccines brings hope to limit the spread of SARS-CoV-2, while the immunogenicity of the vaccines is waning over time. Whether a booster dose of vaccine is needed has become a widely controversial issue. However, no published meta-analysis has focused on the issue. Therefore, this study assessed the immunogenicity and safety of the different combinations of prime-boost vaccinations. Electronic databases including PubMed, the Cochrane Library, Embase, medRxiv, Wanfang and CNKI were used to retrieve the original studies. A total of 28 studies, 9 combinations of prime-boost vaccinations and 5870 subjects were included in the meta-analysis, and random effect models were used to estimate pooled immunogenicity and safety. The immunity against COVID-19 after the prime vaccination waned over time, especially in the populations primed with inactivated vaccines, in which the seropositive rate of antibodies was only 28% (95% CI: 17-40%). Booster vaccination could significantly increase the antibody responses, and heterologous immunization was more effective than homologous immunization (neutralization titers: 1.65 vs. 1.27; anti-RBD IgG: 1.85 vs. 1.15); in particular, the combination of inactivated-mRNA vaccines had the highest antibody responses (neutralization titers: MRAW = 3.64, 95% CI: 3.54-3.74; anti-RBD IgG: 3.73, 95% CI: 3.59-3.87). Moreover, compared with the initial two doses of vaccines, a booster dose did not induce additional or severe adverse events. The administration of the booster dose effectively recalled specific immune responses to SARS-CoV-2 and increased antibody levels, especially in heterologous immunization. Considering the long-term immunogenicity and vaccine equity, we suggest that now, only individuals primed with inactivated vaccines require a booster dose.
Collapse
Affiliation(s)
- Haoyue Cheng
- Department of Public Health and Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Zhicheng Peng
- Department of Public Health and Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Shuting Si
- Department of Public Health and Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Xialidan Alifu
- Department of Public Health and Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Haibo Zhou
- Department of Public Health and Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Peihan Chi
- Department of Public Health and Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Yan Zhuang
- Department of Public Health and Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Minjia Mo
- Department of Public Health and Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Yunxian Yu
- Department of Public Health and Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health, School of Medicine, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
33
|
Fcγ-Receptor-Based Enzyme-Linked Immunosorbent Assays for Sensitive, Specific, and Persistent Detection of Anti-SARS-CoV-2 Nucleocapsid Protein IgG Antibodies in Human Sera. J Clin Microbiol 2022; 60:e0007522. [PMID: 35574677 PMCID: PMC9199419 DOI: 10.1128/jcm.00075-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sensitive and specific serological tests are mandatory for epidemiological studies evaluating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) prevalence as well as coronavirus disease 2019 (COVID-19) morbidity and mortality rates. The accuracy of results is challenged by antibody waning after convalescence and by cross-reactivity induced by previous infections with other pathogens. By employing a patented platform technology based on capturing antigen-antibody complexes with a solid-phase-bound Fcγ receptor (FcγR) and truncated nucleocapsid protein as the antigen, two SARS-CoV-2 IgG enzyme-linked immunosorbent assays (ELISAs), featuring different serum and antigen dilutions, were developed. Validation was performed using a serum panel comprising 213 longitudinal samples from 35 COVID-19 patients and a negative-control panel consisting of 790 pre-COVID-19 samples from different regions of the world. While both assays show similar diagnostic sensitivities in the early convalescent phase, ELISA 2 (featuring a higher serum concentration) enables SARS-CoV-2 IgG antibody detection for a significantly longer time postinfection (≥15 months). Correspondingly, analytical sensitivity referenced to indirect immunofluorescence testing (IIFT) is significantly higher for ELISA 2 in samples with a titer of ≤1:640; for high-titer samples, a prozone effect is observed for ELISA 2. The specificities of both ELISAs were excellent not only for pre-COVID-19 serum samples from Europe, Asia, and South America but also for several challenging African sample panels. The SARS-CoV-2 IgG FcγR ELISAs, methodically combining antigen-antibody binding in solution and isotype-specific detection of immune complexes, are valuable tools for seroprevalence studies requiring the (long-term) detection of anti-SARS-CoV-2 IgG antibodies in populations with a challenging immunological background and/or in which spike-protein-based vaccine programs have been rolled out.
Collapse
|
34
|
Kurra NC, Sriram K, Gandrakota N, Nagarajan JS, Khasnavis S, Ramakrishnan M, Dalal S, Irfan SA, Khan S, JK H, Patel D, Samudrala G. Frontliners on the Move: A Quantitative Analysis of the Prevalence of COVID-19 Reinfection Among Healthcare Workers. Cureus 2022; 14:e24652. [PMID: 35663721 PMCID: PMC9155986 DOI: 10.7759/cureus.24652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/01/2022] [Indexed: 11/05/2022] Open
Abstract
This study was conducted to review relevant articles and demonstrate the prevalence of coronavirus disease 2019 (COVID-19) reinfection among healthcare workers (HCWs). A systemic search was conducted on PubMed and Medline from their inception to July 17, 2021. All statistical analyses were conducted using ReviewManager 5.4.1. Studies meeting the following inclusion criteria were selected: (a) articles having HCWs with COVID-19; (b) studies describing reinfection of COVID-19; and (c) articles having a defined number of patients and controls. Three studies were selected for meta-analysis. The Newcastle-Ottawa Scale (NOS) was used to assess the quality of the cohort studies. NOS scores of 1-5 were considered high risk for bias, scores of 6-7 were deemed moderate, and scores >7 were considered low risk for bias. A random-effect model was used when heterogeneity was seen to pool the studies, and the results were reported in inverse variance (IV) and corresponding 95% confidence interval (CI). Pooled prevalence of reinfection of COVID-19 in HCWs was 3% (OR: 0.03 [-0.04, 0.01]; p=0.44; I2=4%). A non-significant prevalence was found among the healthcare professionals in terms of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) reinfection in Europe. The preformed antibodies were protective against reinfection. However, the waning of antibodies with respect to time was evident, varying differently in different individuals, thereby resulting in reinfection.
Collapse
|
35
|
Burhan E, Rachmadi RA. Omicron surge and the future of COVID-19 vaccinations. MEDICAL JOURNAL OF INDONESIA 2022. [DOI: 10.13181/mji.bc.226066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
The global surge of Omicron has caused significant concern. Omicron has caused new wave of infections in nations with adequate vaccine coverage. Omicron has around 30 mutations that are similar to the previous variant of concerns, possibly decreasing vaccine effectiveness (VE). Hence, the emergence of Omicron is predicted to be a significant public health challenge and may alter the future of COVID-19 vaccinations. Furthermore, other issues may affect vaccine policy in the future such as the never-ending vaccine inequity, waning immunity of current COVID-19 vaccines, decreasing VE against new emerging strains, and new findings regarding hybrid immunity. This literature review aimed to explore the possible steps forward using the most updated knowledge on COVID-19 vaccines and Omicron.
Collapse
|
36
|
Keshavarz B, Richards NE, Workman LJ, Patel J, Muehling LM, Canderan G, Murphy DD, Brovero SG, Ailsworth SM, Eschenbacher WH, McGowan EC, Mann BJ, Nelson MR, Kadl A, Woodfolk JA, Platts-Mills TAE, Wilson JM. Trajectory of IgG to SARS-CoV-2 After Vaccination With BNT162b2 or mRNA-1273 in an Employee Cohort and Comparison With Natural Infection. Front Immunol 2022; 13:850987. [PMID: 35386716 PMCID: PMC8978955 DOI: 10.3389/fimmu.2022.850987] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/28/2022] [Indexed: 01/14/2023] Open
Abstract
Three COVID-19 vaccines have received FDA-authorization and are in use in the United States, but there is limited head-to-head data on the durability of the immune response elicited by these vaccines. Using a quantitative assay we studied binding IgG antibodies elicited by BNT162b2, mRNA-1273 or Ad26.COV2.S in an employee cohort over a span out to 10 months. Age and sex were explored as response modifiers. Of 234 subjects in the vaccine cohort, 114 received BNT162b2, 114 received mRNA-1273 and six received Ad26.COV2.S. IgG levels measured between seven to 20 days after the second vaccination were similar in recipients of BNT162b2 and mRNA-127 and were ~50-fold higher than in recipients of Ad26.COV2.S. However, by day 21 and at later time points IgG levels elicited by BNT162b2 were lower than mRNA-1273. Accordingly, the IgG decay curve was steeper for BNT162b2 than mRNA-1273. Age was a significant modifier of IgG levels in recipients of BNT162b2, but not mRNA-1273. After six months, IgG levels elicited by BNT162b2, but not mRNA-1273, were lower than IgG levels in patients who had been hospitalized with COVID-19 six months earlier. Similar findings were observed when comparing vaccine-elicited antibodies with steady-state IgG targeting seasonal human coronaviruses. Differential IgG decay could contribute to differences observed in clinical protection over time between BNT162b2 and mRNA-1273.
Collapse
Affiliation(s)
- Behnam Keshavarz
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Nathan E Richards
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Lisa J Workman
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Jaimin Patel
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Lyndsey M Muehling
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Glenda Canderan
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Deborah D Murphy
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Savannah G Brovero
- Division of Infectious Disease and International Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Samuel M Ailsworth
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Will H Eschenbacher
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Emily C McGowan
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Barbara J Mann
- Division of Infectious Disease and International Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Michael R Nelson
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Alexandra Kadl
- Division of Pulmonary and Critical Care, Department of Medicine, University of Virginia, Charlottesville, VA, United States.,Department of Pharmacology, University of Virginia, Charlottesville, VA, United States
| | - Judith A Woodfolk
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Thomas A E Platts-Mills
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Jeffrey M Wilson
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
37
|
Hatmal MM, Al-Hatamleh MAI, Olaimat AN, Mohamud R, Fawaz M, Kateeb ET, Alkhairy OK, Tayyem R, Lounis M, Al-Raeei M, Dana RK, Al-Ameer HJ, Taha MO, Bindayna KM. Reported Adverse Effects and Attitudes among Arab Populations Following COVID-19 Vaccination: A Large-Scale Multinational Study Implementing Machine Learning Tools in Predicting Post-Vaccination Adverse Effects Based on Predisposing Factors. Vaccines (Basel) 2022; 10:366. [PMID: 35334998 PMCID: PMC8955470 DOI: 10.3390/vaccines10030366] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/04/2023] Open
Abstract
Background: The unprecedented global spread of coronavirus disease 2019 (COVID-19) has imposed huge challenges on the healthcare facilities, and impacted every aspect of life. This has led to the development of several vaccines against COVID-19 within one year. This study aimed to assess the attitudes and the side effects among Arab communities after receiving a COVID-19 vaccine and use of machine learning (ML) tools to predict post-vaccination side effects based on predisposing factors. Methods: An online-based multinational survey was carried out via social media platforms from 14 June to 31 August 2021, targeting individuals who received at least one dose of a COVID-19 vaccine from 22 Arab countries. Descriptive statistics, correlation, and chi-square tests were used to analyze the data. Moreover, extensive ML tools were utilized to predict 30 post vaccination adverse effects and their severity based on 15 predisposing factors. The importance of distinct predisposing factors in predicting particular side effects was determined using global feature importance employing gradient boost as AutoML. Results: A total of 10,064 participants from 19 Arab countries were included in this study. Around 56% were female and 59% were aged from 20 to 39 years old. A high rate of vaccine hesitancy (51%) was reported among participants. Almost 88% of the participants were vaccinated with one of three COVID-19 vaccines, including Pfizer-BioNTech (52.8%), AstraZeneca (20.7%), and Sinopharm (14.2%). About 72% of participants experienced post-vaccination side effects. This study reports statistically significant associations (p < 0.01) between various predisposing factors and post-vaccinations side effects. In terms of predicting post-vaccination side effects, gradient boost, random forest, and XGBoost outperformed other ML methods. The most important predisposing factors for predicting certain side effects (i.e., tiredness, fever, headache, injection site pain and swelling, myalgia, and sleepiness and laziness) were revealed to be the number of doses, gender, type of vaccine, age, and hesitancy to receive a COVID-19 vaccine. Conclusions: The reported side effects following COVID-19 vaccination among Arab populations are usually non-life-threatening; flu-like symptoms and injection site pain. Certain predisposing factors have greater weight and importance as input data in predicting post-vaccination side effects. Based on the most significant input data, ML can also be used to predict these side effects; people with certain predicted side effects may require additional medical attention, or possibly hospitalization.
Collapse
Affiliation(s)
- Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan
| | - Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia; (M.A.I.A.-H.); (R.M.)
| | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia; (M.A.I.A.-H.); (R.M.)
| | - Mirna Fawaz
- Nursing Department, Faculty of Health Sciences, Beirut Arab University, Beirut 1105, Lebanon;
| | - Elham T. Kateeb
- Oral Health Research and Promotion Unit, Faculty of Dentistry, Al-Quds University, Jerusalem 51000, Palestine;
| | - Omar K. Alkhairy
- Department of Pathology and Laboratory Medicine, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, Riyadh 11426, Saudi Arabia;
- King Saud bin Abdulaziz University for Health Sciences, P.O. Box 3660, Riyadh 11481, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), P.O. Box 3660, Riyadh 11481, Saudi Arabia
| | - Reema Tayyem
- Department of Human Nutrition, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Mohamed Lounis
- Department of Agro-Veterinary Science, Faculty of Natural and Life Sciences, University of Ziane Achour, BP 3117, Djelfa 17000, Algeria;
| | - Marwan Al-Raeei
- Faculty of Sciences, Damascus University, Damascus P.O. Box 30621, Syria;
| | - Rasheed K. Dana
- Faculty of Medicine, Mansoura University, Mansoura, Dakahlia 35516, Egypt;
| | - Hamzeh J. Al-Ameer
- Department of Biology and Biotechnology, Faculty of Science, American University of Madaba, P.O. Box 99, Madaba 17110, Jordan;
| | - Mutasem O. Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan, Amman 11942, Jordan;
| | - Khalid M. Bindayna
- Department of Microbiology, Immunology and Infectious Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama 329, Bahrain
| |
Collapse
|
38
|
Lau CS, Phua SK, Liang YL, Oh MLH, Aw TC. SARS-CoV-2 Spike and Neutralizing Antibody Kinetics 90 Days after Three Doses of BNT162b2 mRNA COVID-19 Vaccine in Singapore. Vaccines (Basel) 2022; 10:331. [PMID: 35214789 PMCID: PMC8879250 DOI: 10.3390/vaccines10020331] [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: 01/28/2022] [Revised: 02/09/2022] [Accepted: 02/12/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND We evaluated the post-booster (BNT162b2) antibody responses in Singapore. METHODS Participants (n = 43) were tested pre-booster and 20/30/60/90 days post-booster. Participants were boosted 120-240 days (mean 214 days) after their second dose and had no history or serologic evidence of prior COVID-19 infection; all participants had undetectable SARS-CoV-2 nucleocapsid antibodies throughout the study. Total nucleocapsid and spike antibodies (S-Ab) were assessed on the Roche Elecsys e802 and neutralizing antibody (N-Ab) on the Snibe quantitative N-Ab assay. RESULTS Pre-booster median S-Ab/N-Ab titers were 829 BAU/mL/0.83 µg/mL; 2 participants were below manufacturer's N-Ab cut-offs of 0.3 µg/mL (0.192 and 0.229). Both S-Ab and N-Ab titers peaked at 30 days post-booster (median S-Ab 25,220 BAU/mL and N-Ab 30.3 µg/mL) at 30-37× pre-booster median levels. These peak post-booster S-Ab/N-Ab titers were 11× (25,220 vs. 2235 BAU/mL) and 9× (30.3 vs. 3.52 µg/mL) higher than the previously reported peak post-second dose levels. Antibody titers declined to 12,315 BAU/mL (51% decrease) and 14.3 µg/mL (53% decrease) 90 days post-booster. Non-linear regression estimates for S-Ab/N-Ab half-lives were 44/58 days. At 180 days post-booster, S-Ab/N-Ab are estimated to be 2671 BAU/mL/4.83 µg/mL. CONCLUSIONS Both S-Ab and N-Ab show a good response following post-booster vaccination, with half-lives that may provide a prolonged antibody response.
Collapse
Affiliation(s)
- Chin Shern Lau
- Department of Laboratory Medicine, Changi General Hospital, Singapore 529889, Singapore; (S.K.P.); (Y.L.L.); (T.C.A.)
| | - Soon Kieng Phua
- Department of Laboratory Medicine, Changi General Hospital, Singapore 529889, Singapore; (S.K.P.); (Y.L.L.); (T.C.A.)
| | - Ya Li Liang
- Department of Laboratory Medicine, Changi General Hospital, Singapore 529889, Singapore; (S.K.P.); (Y.L.L.); (T.C.A.)
| | - May Lin Helen Oh
- Department of Infectious Diseases, Changi General Hospital, Singapore 529889, Singapore;
| | - Tar Choon Aw
- Department of Laboratory Medicine, Changi General Hospital, Singapore 529889, Singapore; (S.K.P.); (Y.L.L.); (T.C.A.)
- Department of Medicine, National University of Singapore, Singapore 119077, Singapore
- Academic Pathology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
39
|
Rabiu M, Iyaniwura SA. Assessing the potential impact of immunity waning on the dynamics of COVID-19 in South Africa: an endemic model of COVID-19. NONLINEAR DYNAMICS 2022; 109:203-223. [PMID: 35095199 PMCID: PMC8788409 DOI: 10.1007/s11071-022-07225-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/09/2022] [Indexed: 05/05/2023]
Abstract
We developed an endemic model of COVID-19 to assess the impact of vaccination and immunity waning on the dynamics of the disease. Our model exhibits the phenomenon of backward bifurcation and bi-stability, where a stable disease-free equilibrium coexists with a stable endemic equilibrium. The epidemiological implication of this is that the control reproduction number being less than unity is no longer sufficient to guarantee disease eradication. We showed that this phenomenon could be eliminated by either increasing the vaccine efficacy or by reducing the disease transmission rate (adhering to non-pharmaceutical interventions). Furthermore, we numerically investigated the impacts of vaccination and waning of both vaccine-induced immunity and post-recovery immunity on the disease dynamics. Our simulation results show that the waning of vaccine-induced immunity has more effect on the disease dynamics relative to post-recovery immunity waning and suggests that more emphasis should be on reducing the waning of vaccine-induced immunity to eradicate COVID-19.
Collapse
Affiliation(s)
- Musa Rabiu
- School of Mathematics, Statistics & Computer Science, University of KwaZulu-Natal, Durban, South Africa
| | - Sarafa A Iyaniwura
- Department of Mathematics and Institute of Applied Mathematics, University of British Columbia, Vancouver, BC Canada
| |
Collapse
|
40
|
Immunity after COVID-19 and vaccination: follow-up study over 1 year among medical personnel. Infection 2021; 50:439-446. [PMID: 34562263 PMCID: PMC8475821 DOI: 10.1007/s15010-021-01703-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/18/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND The long-term course of immunity among individuals with a history of COVID-19, in particular among those who received a booster vaccination, has not been well defined so far. METHODS SARS-CoV-2-specific antibody levels were measured by ELISA over 1 year among 136 health care workers infected during the first COVID-19 wave and in a subgroup after booster vaccination approximately 1 year later. Furthermore, spike-protein-reactive memory T cells were quantified approximately 7 months after the infection and after booster vaccination. Thirty healthy individuals without history of COVID-19 who were routinely vaccinated served as controls. RESULTS Levels of SARS-CoV-2-specific IgM- and IgA-antibodies showed a rapid decay over time, whereas IgG-antibody levels decreased more slowly. Among individuals with history of COVID-19, booster vaccination induced very high IgG- and to a lesser degree IgA-antibodies. Antibody levels were significantly higher after booster vaccination than after recovery from COVID-19. After vaccination with a two-dose schedule, healthy control subjects developed similar antibody levels as compared to individuals with history of COVID-19 and booster vaccination. SARS-CoV-2-specific memory T cell counts did not correlate with antibody levels. None of the study participants suffered from a reinfection. CONCLUSIONS Booster vaccination induces high antibody levels in individuals with a history of COVID-19 that exceeds by far levels observed after recovery. SARS-CoV-2-specific antibody levels of similar magnitude were achieved in healthy, COVID-19-naïve individuals after routine two-dose vaccination.
Collapse
|