1
|
Novelli S, Reinkemeyer C, Bulaev D, O’Sullivan MP, Snoeck CJ, Rauschenberger A, Manto C, Kolodkin A, Ghosh S, Satagopam V, le Chenadec J, Barthelemy K, Priet S, de Lamballerie X, Wieser A, Kroidl I, Vaillant M, Meyer L, Hoelscher M, Castelletti N, Krüger R, Warszawski J. Waning of anti-SARS-CoV-2 antibodies after the first wave of the COVID-19 pandemic in 2020: A 12-month-evaluation in three population-based European studies. PLoS One 2025; 20:e0320196. [PMID: 40344145 PMCID: PMC12063904 DOI: 10.1371/journal.pone.0320196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/15/2025] [Indexed: 05/11/2025] Open
Abstract
OBJECTIVES We described waning in anti-SARS-CoV-2 IgG in adult general populations infected during the first wave of the COVID-19 pandemic in 2020 across three European countries. METHODS Coordinated analyses were conducted separately in three population-based cohorts with complementary follow-up schedules: the KoCo19 (Germany), EpiCov (France), and CON-VINCE (Luxembourg) cohorts. Serological follow-up was based on the anti-SARS-CoV-2 ELISA-S IgG (Euroimmun) assay. We selected all adults aged 18-79 who had a positive serology (IgG optical density (OD) ratio ≥1.1) between February and July 2020, and at least one subsequent IgG measurement within the following 12 months, while still non-vaccinated. RESULTS The proportion of seroreversion was 0% within the four first months, based on Koco19 data (n = 65 participants). In the longer term, 31.3% of participants had seroreverted at 6 months (95%CI: 24.4-39.1) (based on EpiCov data, n = 599), 31.3% (95%CI: 11.0-58.7) at 12 months (based on CON-VINCE data, n = 16). From EpiCov data, both baseline low IgG levels and seroneutralization negativity remained predictive of seroreversion in multivariable analysis. CONCLUSION From population-based cohorts, anti-S IgG levels remained stable during the first 4 months following SARS-CoV-2 infection. Most of the decay occurred afterward; nearly one-third of people seroreverted 6 and 12 months later. Low IgG levels and seroneutralization negativity were independent predictors of seroreversion.
Collapse
Affiliation(s)
- Sophie Novelli
- Université Paris-Saclay, UVSQ, Inserm, CESP, APHP Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Christina Reinkemeyer
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research, Munich, Germany
| | - Dmitry Bulaev
- Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | | | | | - Armin Rauschenberger
- Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Carmelite Manto
- Université Paris-Saclay, UVSQ, Inserm, CESP, Le Kremlin-Bicêtre, France
| | | | - Soumyabrata Ghosh
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Venkata Satagopam
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | | | - Karine Barthelemy
- Unité des Virus Emergents, UVE, Aix Marseille Université, Inserm, France
| | - Stephane Priet
- Unité des Virus Emergents, UVE, Aix Marseille Université, Inserm, France
| | | | - Andreas Wieser
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
- Max Von Pettenkofer Institute, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Inge Kroidl
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| | | | - Laurence Meyer
- Université Paris-Saclay, UVSQ, Inserm, CESP, APHP Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Michael Hoelscher
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
- Center for International Health (CIH), University Hospital, LMU Munich, Munich, Germany
- Unit Global Health, Helmholtz Zentrum München, German Research Centre for Environmental Health (HMGU), Neuherberg, Germany
| | - Noemi Castelletti
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
- Institute of Radiation Medicine, Helmholtz Zentrum München, Neuherberg, Germany
| | - Rejko Krüger
- Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), Strassen, Luxembourg
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Josiane Warszawski
- Université Paris-Saclay, UVSQ, Inserm, CESP, APHP Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | | |
Collapse
|
2
|
Dhawan M, Thakur N, Sharma M, Rabaan AA. The comprehensive insights into the B-cells-mediated immune response against COVID-19 infection amid the ongoing evolution of SARS-CoV-2. Biomed Pharmacother 2025; 185:117936. [PMID: 40056829 DOI: 10.1016/j.biopha.2025.117936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 02/08/2025] [Accepted: 02/20/2025] [Indexed: 03/10/2025] Open
Abstract
The antibody-mediated immune response is crucial for the development of protective immunity against SARS-CoV-2, the virus responsible for the COVID-19 pandemic. Understanding the interaction between SARS-CoV-2 and the immune system is critical because new variants emerge as a result of the virus's ongoing evolution. Understanding the function of B cells in the SARS-CoV-2 infection process is critical for developing effective and long-lasting vaccines against this virus. Triggered by the innate immune response, B cells transform into memory B cells (MBCs). It is fascinating to observe how MBCs provide enduring immune defence, not only eradicating the infection but also safeguarding against future reinfection. If there is a lack of B cell activation or if the B cells are not functioning properly, it can lead to a serious manifestation of the disease and make immunisation less effective. Individuals with disruptions in the B cells have shown increased production of cytokines and chemokines, resulting in a poor prognosis for the disease. Therefore, we have developed an updated review article to gain insight into the involvement of B cells in SARS-CoV-2 infection. The discussion has covered the generation, functioning, and dynamics of neutralising antibodies (nAbs). Furthermore, we have emphasised immunotherapeutics that rely on nAbs.
Collapse
Affiliation(s)
- Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, Punjab 141004, India; Trafford College, Altrincham, Altrincham, Manchester WA14 5PQ, UK.
| | - Nanamika Thakur
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, India
| | - Manish Sharma
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, India
| | - Ali A Rabaan
- Research Center, Dr. Sulaiman Alhabib Medical Group, Riyadh 13328, Saudi Arabia; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan.
| |
Collapse
|
3
|
Nouanesengsy A, Semesi A, Quach K, Ivanochko D, Byrne W, Hwang M, La Neve MR, Leon-Ponte M, Litosh A, Wisener N, Adeli K, Campigotto A, Grunebaum E, McGeer A, Moraes TJ, Sepiashvili L, Upton J, Julien JP, Allen U. Persistence and decay of neutralizing antibody responses elicited by SARS-CoV-2 infection and hybrid immunity in a Canadian cohort. Microbiol Spectr 2025; 13:e0133324. [PMID: 39969224 PMCID: PMC11960127 DOI: 10.1128/spectrum.01333-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
A major challenge with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has been assessing the intensity, dynamics, and determinants of the antibody responses after infection and/or vaccination. Therefore, we aimed to characterize the longitudinal dynamics of the antibody responses among naturally infected individuals and individuals who achieved hybrid immunity in a large Canadian cohort. We demonstrate that anti-Spike IgGs and neutralizing antibody dynamics vary greatly among individuals with COVID-19, in peak antibody levels, rate of waning, and longevity of the antibody response. Additionally, we found an association between robust antibody responses and individuals with severe COVID-19 clinical symptoms during the first-month post-symptom onset. For individuals who achieved hybrid immunity, a robust increase in anti-S1 IgGs and neutralizing antibodies followed the first vaccination dose; however, there was a minimal increase in the anti-S1 IgGs and neutralizing antibody titers after administration of the second dose of the vaccine. Furthermore, neutralizing antibodies elicited by the wild-type virus alone were largely ineffective against emerging variants of concern in our natural infection-only cohort, in contrast to a much broader and more robust neutralization profile observed in individuals who achieved hybrid immunity. Our findings emphasize the need for global SARS-CoV-2 vaccination efforts to further sustain protective immune responses required to minimize viral spread and disease severity in the population. As SARS-CoV-2 variants continue to emerge, understanding the interplay between previous infections, vaccine durability, and virus evolution will be critical for guiding ongoing vaccination strategies. IMPORTANCE A major challenge with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has been assessing the intensity, dynamics, and determinants of the antibody response after infection and/or vaccination. Our paper addresses this in a large Canadian cohort with antibody responses that were generated by natural infection as well as vaccine in some persons studied.
Collapse
Affiliation(s)
- Amy Nouanesengsy
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Anthony Semesi
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
| | - Kim Quach
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Danton Ivanochko
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
| | - Walter Byrne
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Matthew Hwang
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maria-Rosa La Neve
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Matilde Leon-Ponte
- Division of Allergy and Immunology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alice Litosh
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nicole Wisener
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Khosrow Adeli
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Aaron Campigotto
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Eyal Grunebaum
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
| | - Allison McGeer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tunenbaum Research Institute at Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Theo J. Moraes
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lusia Sepiashvili
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Julia Upton
- Division of Allergy and Immunology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Upton Allen
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
- Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Zheng X, Chen Q, Liao Q, Zhang X. Tracking the evolution of serum antibody levels and influencing factors post-SARS-CoV-2 infection among community residents in Fuzhou City. Front Immunol 2025; 16:1533102. [PMID: 40230855 PMCID: PMC11994895 DOI: 10.3389/fimmu.2025.1533102] [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: 11/23/2024] [Accepted: 03/12/2025] [Indexed: 04/16/2025] Open
Abstract
Objective To track the level of serum antibodies in Fuzhou residents and analyze the possible influencing factors of serum antibodies, so as to provide a scientific basis for the adjustment of population immunity and prevention and control strategies. Methods Residents in the Fuzhou community who had symptoms of covid-19 infection or who had tested positive for nucleic acid or antigen since December 2022 were selected for the questionnaire survey and their sera were collected to analyze the trend of antibody changes, the antibody level was divided into high antibody group and low antibody group according to the literature data. The possible influencing factors of serum antibody level was analyzed by multivariate logistic regression model. Results A total of 2,521 Fuzhou residents were adopted in the study, including 223 in the high antibody group and 194 in the low antibody group. A univariate analysis showed that, there were significant differences in age (Z=-4.028, P<0.00), occupation (χ2 = 18.591, P=0.005), typical symptoms after the first infection (χ2 = 9.784, P=0.002), history of surgery (χ2 = 29.542, P<0.001), symptoms lasting more than 2 weeks after the first infection (χ2 = 4.887, P=0.027), smoking (χ2 = 18.524, P<0.001) and drinking (χ2 = 19.578, P<0.001) between the high antibody group and the low antibody group. Multivariate regression models show that, age (OR= 1.011, 95%CI: 1.002~1.020, P=0.017), history of surgery (OR=4.956,95%CI: 2.606~9.423, P<0.001),smoking (OR=2.089, 95%CI: 1.002~4.355, P=0.049), drinking (OR=2.214, 95%CI: 1.066~4.600, P=0.033) were the risk factors affecting antibody level. Typical symptoms after the first infection (OR=0.224, 95%CI: 0.086~0.579, P=0.002) and symptoms lasting more than 2 weeks after the first infection (OR=0.432, 95%CI: 0.258~0.723, P=0.001) were protective factors. By observing the trend of antibody changes in 3, 6 and 9 months, we found that the level of IgG antibody showed a decreasing trend. Conclusions The high level of protection was more likely to occur in young adults, people without operation history, people without smoking history, people without drinking history, people with typical symptoms after the first infection and symptoms lasting more than 2 weeks after the first infection. The level of IgG antibody was decreased in general, so it is necessary to strengthen immunization.
Collapse
Affiliation(s)
- Xiaoyan Zheng
- The Affiliated Fuzhou Center for Disease Control and Prevention of Fujian Medical University, Fuzhou, China
- The School of Public Health, Fujian Medical University, Fuzhou, China
| | - Qingquan Chen
- The Affiliated Fuzhou Center for Disease Control and Prevention of Fujian Medical University, Fuzhou, China
- The School of Public Health, Fujian Medical University, Fuzhou, China
| | - Qiangbing Liao
- The Affiliated Fuzhou Center for Disease Control and Prevention of Fujian Medical University, Fuzhou, China
- The School of Public Health, Fujian Medical University, Fuzhou, China
| | - Xiaoyang Zhang
- The Affiliated Fuzhou Center for Disease Control and Prevention of Fujian Medical University, Fuzhou, China
- The School of Public Health, Fujian Medical University, Fuzhou, China
| |
Collapse
|
5
|
Fang JL, Shrestha L, Beland FA. Flow cytometric analysis of the SARS coronavirus 2 antibodies in human plasma. Sci Rep 2025; 15:10300. [PMID: 40133428 PMCID: PMC11937374 DOI: 10.1038/s41598-025-92389-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
COVID-19 is an infectious disease caused by the severe acute respiratory syndrome coronavirus (SARS-CoV-2). Anti-SARS-CoV-2 antibodies can provide information on patient immunity, identify asymptomatic patients, and track the spread of COVID-19. Efforts have been made to develop methods to detect anti-SARS-CoV-2 antibodies in humans. Here, we describe a flow cytometric assay for the simultaneous detection of anti-SARS-CoV-2 IgG and IgM in human plasma. To assess the antibody response against the different SARS-CoV-2 structural proteins, five viral recombinant proteins, including spike protein subunit 1 (S1), N-terminal domain of S1 (S1A), spike receptor-binding domain (RBD), spike protein subunit 2 (S2), and nucleocapsid protein (N), were generated. A comparison of the antibody profiles detected by the assay with plasma from 100 healthy blood donors collected prior to the COVID-19 pandemic and plasma from 100 virologically confirmed COVID-19 patients demonstrated a clear discrimination between the two groups. Among the COVID-19 patients, the antibody responses for the viral proteins, as determined by their prevalence, were anti-RBD IgG = anti-N IgG > anti-S1 IgG > anti-S1A IgG > anti-S2 IgG, and anti-RBD IgM > anti-S1 IgM > anti-N IgM > anti-S2 IgM. The prevalence of anti-SARS-CoV-2 IgG and IgM was not associated with sex, age, race, days after the onset of symptoms, or severity of illness, except for a higher prevalence of anti-S2 IgG being observed in men than in women. The levels of anti-RBD IgG were higher in patients 65 years and older and in patients who had severe symptoms. Similarly, patients who had severe symptoms exhibited higher levels of anti-S1 and anti-S1A IgG than patients who had mild or moderate symptoms. The levels of anti-RBD IgM tended to be higher in men but did not differ among age, race, days after the onset of symptoms, or severity of illness. Our study indicates that the flow cytometric assay, especially using RBD as target antigen, can be used to detect simultaneously anti-SARS-CoV-2 IgG and IgM antibodies in human plasma.
Collapse
Affiliation(s)
- Jia-Long Fang
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA.
| | - Leeza Shrestha
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| |
Collapse
|
6
|
Honda-Okubo Y, Sajkov D, Wauchope B, Turner JV, Vote B, Antipov A, André G, Lebedin Y, Petrovsky N. Immunogenicity and safety study of a single dose of SpikoGen® vaccine as a heterologous or homologous intramuscular booster following a primary course of mRNA, adenoviral vector or recombinant protein COVID-19 vaccine in ambulatory adults. Vaccine 2025; 49:126744. [PMID: 39914274 DOI: 10.1016/j.vaccine.2025.126744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 01/13/2025] [Accepted: 01/13/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND SpikoGen® is a subunit recombinant Wuhan spike protein produced in insect cells and formulated with Advax-CpG55.2™ adjuvant. It is approved for adult and pediatric use in the Middle East. This study tested the safety and immunogenicity of SpikoGen® as a 3rd, 4th or 5th dose booster following a primary immunisation course of mRNA, adenovirus or SpikoGen® vaccine. METHODS The trial recruited participants who had received a previous doses of COVID-19 vaccine more than 3 months prior. Each received a single intramuscular booster dose of SpikoGen® vaccine. Spike and nuclear protein antibody levels were measured at 1 and 3 months post-booster, together with collection of data on SARS-CoV-2 breakthrough infections and symptoms of long COVID. RESULTS One-month post-booster, anti-spike IgG, sVNT, and pVNT levels were increased in all groups and there was ∼4-fold neutralizing antibodies against the heterologous Omicron BA.2 and BA.4/5 strains. The SpikoGen®-prime group had the highest levels of anti-spike IgG3, consistent with the Advax-CpG adjuvant driving IgG3 induction. There was no effect of age on the vaccine response. The booster dose was well tolerated with no vaccine-associated serious adverse events. Nine participants (9/74, 12.2 %) had a breakthrough SARS-CoV-2 infection between 2 weeks and 3 months post-booster. No long COVID was observed after breakthrough infections. Breakthrough infection was negatively correlated with baseline anti-nuclear protein IgG seropositivity. CONCLUSION A single SpikoGen® booster was well tolerated and stimulated cross- antibody responses against Omicron variants, regardless of the primary vaccine course received. With SARS-CoV-2 variants continuing to evolve, ongoing research is needed into optimum booster strategies. CLINICALTRIALS gov registration. NCT05542862.
Collapse
MESH Headings
- Humans
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/adverse effects
- Immunization, Secondary/methods
- Male
- Adult
- Female
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Spike Glycoprotein, Coronavirus/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Middle Aged
- SARS-CoV-2/immunology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Injections, Intramuscular
- Immunogenicity, Vaccine
- Adenoviridae/genetics
- Young Adult
- Immunoglobulin G/blood
- Genetic Vectors
Collapse
Affiliation(s)
- Yoshikazu Honda-Okubo
- Vaxine Pty Ltd, Warradale, Adelaide, SA 5046, Australia; Australian Respiratory and Sleep Medicine Institute Ltd, Adelaide, SA 5042, Australia
| | - Dimitar Sajkov
- Australian Respiratory and Sleep Medicine Institute Ltd, Adelaide, SA 5042, Australia
| | - Bruce Wauchope
- Bedford Clinic, South Road, Adelaide, SA 5039, Australia
| | - Joseph V Turner
- School of Rural Medicine, University of New England, Armidale, NSW 2351, Australia
| | - Brendan Vote
- Tasmanian Eye Institute Ltd, Launceston, Tasmania 7250, Australia
| | - Anna Antipov
- Vaxine Pty Ltd, Warradale, Adelaide, SA 5046, Australia
| | - Greiciely André
- Vaxine Pty Ltd, Warradale, Adelaide, SA 5046, Australia; Australian Respiratory and Sleep Medicine Institute Ltd, Adelaide, SA 5042, Australia
| | | | - Nikolai Petrovsky
- Vaxine Pty Ltd, Warradale, Adelaide, SA 5046, Australia; Australian Respiratory and Sleep Medicine Institute Ltd, Adelaide, SA 5042, Australia.
| |
Collapse
|
7
|
Kasbergen LMR, de Bruin E, Chandler F, Sigfrid L, Chan XHS, Hookham L, Wei J, Chen S, GeurtsvanKessel CH, Scherbeijn S, Charrel RN, Ayhan N, Lee JL, Corman VM, Reusken C, Loens K, Popescu CP, Lupse M, Briciu V, Văsieşiu AM, Pipero P, Harxhi A, Puca E, Ponosheci Biçaku A, Travar M, Ostojić M, Baljic R, Arapović J, Ledina D, Cekinović Grbeša Đ, Čabraja I, Kurolt IC, Halichidis S, Birlutiu V, Dumitru IM, Moroti R, Barac A, Stevanovic G, Pyrpasopoulou A, Koulouras V, Betica Radić L, Papanikolaou MN, Roilides E, Markotić A, Galal U, Denis E, Goodwin L, Turtle L, Florescu SA, Ramadani H, Goossens H, Ieven M, Drosten C, Horby PW, Sikkema RS, Koopmans MPG. Multi-antigen serology and a diagnostic algorithm for the detection of arbovirus infections as novel tools for arbovirus preparedness in southeast Europe (MERMAIDS-ARBO): a prospective observational study. THE LANCET. INFECTIOUS DISEASES 2025:S1473-3099(24)00654-6. [PMID: 39987930 DOI: 10.1016/s1473-3099(24)00654-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/06/2024] [Accepted: 09/24/2024] [Indexed: 02/25/2025]
Abstract
BACKGROUND Arboviruses are increasingly affecting Europe, partly due to the effects of climate change. This increase in range and impact emphasises the need to improve preparedness for emerging arboviral infections that often co-circulate and might have overlapping clinical syndromes. We aimed to strengthen surveillance networks for four clinically relevant arboviruses in southeast Europe. METHODS This study reports an in-depth analysis of the MERMAIDS-ARBO prospective observational study in adults (ie, aged ≥18 years) hospitalised with an arbovirus-compatible disease syndrome in 21 hospitals in seven countries in southeast Europe over four arbovirus seasons (May 1-Oct 31, 2016-19) to obtain arbovirus prevalence outcomes. The main objectives of the MERMAIDS-ARBO study, describing the clinical management and outcomes of four arboviruses endemic to southeast Europe, including Crimean-Congo haemorrhagic fever virus (CCHFV), tick-borne encephalitis virus (TBEV), Toscana virus, and West Nile virus (WNV), are reported elsewhere. In this analysis, given the challenges associated with arbovirus diagnostics, we developed a diagnostic algorithm accounting for serology outcomes and sample timing to study arbovirus prevalence in southeast Europe. Serum samples were collected on days 0, 7, 28, and 60 after hospital admission and tested for anti-CCHFV IgG and IgM antibodies with ELISAs (confirmed with an indirect immunofluorescence test) and for IgG and IgM antibodies specific to TBEV, Toscana virus, and WNV with custom-printed protein microarrays (confirmed with virus neutralisation tests). All acute-phase samples were tested by PCR for all four viruses. Descriptive analyses were performed for virus-reactive cases by geography and year, and possible factors (eg, age, sex, and insect bites) associated with virus reactivity were assessed. FINDINGS Of 2896 individuals screened, 913 were eligible for inclusion, of whom 863 (514 men, 332 women, and 17 unknown) had samples sent to the study reference laboratories and were included in molecular and serological analyses. Some individuals had insufficient clinical data to be included in the clinical analysis, but met the eligibility criteria for and were included here. Serum sampling was incomplete (eg, samples missing from one or more timepoints or no data on time since symptom onset) for 602 (70%) patients, and the timing of collection was often heterogeneous after symptom onset up to 40 days (average median delay of 5-6 days across all timepoints), affecting the ability to diagnose arbovirus infection by serology. By use of an interpretation table incorporating timing and completeness of sampling, one (<1%) participant had a confirmed recent infection with CCHFV, ten (1%) with TBEV, 40 (5%) with Toscana virus, and 52 (6%) with WNV. Most acute confirmed infections of Toscana virus were found in Albania (25 [63%] of 40), whereas WNV was primarily identified in Romania (36 [69%] of 52). Albania also had the highest overall Toscana virus seropositivity (168 [60%] of 282), mainly explained by patients confirmed to be exposed or previously exposed (104 [62%] of 168). Patients without antibodies to WNV or Toscana virus were significantly younger than patients with antibodies (mean difference -8·48 years [95% CI -12·31 to -4·64] for WNV, and -6·97 years [-9·59 to -4·35] for Toscana virus). We found higher odds of Toscana virus reactivity in men (odds ratio 1·56 [95% CI 1·15 to 2·11]; p=0·0055), WNV reactivity with mosquito bites versus no mosquito bites (2·47 [1·54 to 3·97]; p=0·0002), and TBEV reactivity with tick bites versus no tick bites (2·21 [1·19 to 4·11]; p=0·018). INTERPRETATION This study shows that despite incomplete and heterogeneous data, differential diagnosis of suspected arbovirus infections is possible, and the diagnostic interpretation algorithm we propose could potentially be used to strengthen routine diagnostics in clinical settings in areas at risk for arboviral diseases. Our data highlight potential hotspots for arbovirus surveillance and risk factors associated with these particular arbovirus infections. FUNDING European Commission and Versatile Emerging infectious disease Observatory. TRANSLATIONS For the Greek, Albanian, Romanian, Bosnian, Serbian, and Croatian translation of the summary see Supplementary Materials section.
Collapse
Affiliation(s)
- Louella M R Kasbergen
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands.
| | - Erwin de Bruin
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Felicity Chandler
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Louise Sigfrid
- ERGO, Pandemic Sciences Institute, University of Oxford, Oxford, UK; Policy and Practice Research Group, Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | - Xin Hui S Chan
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Lauren Hookham
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jia Wei
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Siyu Chen
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; High Meadows Environmental Institute, Princeton University, Princeton, NJ, US
| | | | - Sandra Scherbeijn
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Remi N Charrel
- Unite des Virus Emergents, Aix-Marseille Université, Universita di Corsica, IRD 190, Inserm 1207, IRBA, Marseille, France
| | - Nazli Ayhan
- Unite des Virus Emergents, Aix-Marseille Université, Universita di Corsica, IRD 190, Inserm 1207, IRBA, Marseille, France; Centre National de Référence des Arbovirus, Inserm-IRBA, Marseille, France
| | - James L Lee
- ERGO, Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | - Victor M Corman
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; German Center for Infection Research (DZIF), Berlin, Germany
| | - Chantal Reusken
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands; Centre for Infectious Disease Control, National Institute for Public Health and the Environment, RIVML, Bilthoven, Netherlands
| | - Katherine Loens
- Department of Medical Microbiology, University of Antwerp UIA, Antwerp, Belgium
| | - Corneliu Petru Popescu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Dr Victor Babes Clinical Hospital of Infectious and Tropical Diseases, Bucharest, Romania
| | - Mihaela Lupse
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania; The Teaching Hospital for Infectious Diseases, Cluj-Napoca, Romania
| | - Violeta Briciu
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania; The Teaching Hospital for Infectious Diseases, Cluj-Napoca, Romania
| | - Anca Meda Văsieşiu
- Department of Infectious Diseases, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, Târgu Mureş, Romania
| | - Pellumb Pipero
- Department of Infectious Diseases, Mother Teresa University Hospital Center, Tirana, Albania
| | - Arjan Harxhi
- Faculty of Medicine, Medical University of Tirana, Tirana, Albania
| | - Edmond Puca
- Department of Infectious Diseases, Mother Teresa University Hospital Center, Tirana, Albania
| | | | - Maja Travar
- Department of Microbiology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Maja Ostojić
- School of Medicine, University of Mostar, Mostar, Bosnia and Herzegovina
| | - Rusmir Baljic
- Unit for Infectious Disease, Clinical Center of the University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Jurica Arapović
- School of Medicine, University of Mostar, Mostar, Bosnia and Herzegovina; Department of Infectious Diseases, University Clinical Hospital Mostar, Mostar, Bosnia and Herzegovina
| | - Dragan Ledina
- Department of Infectious Diseases, University Hospital Split, Split, Croatia
| | | | - Ivica Čabraja
- Department of Infectious Diseases, Dr Josip Benčević General Hospital, Slavonski Brod, Croatia
| | | | - Stela Halichidis
- Clinical Infectious Diseases Hospital, Constanța, Romania; Faculty of Medicine, Ovidius University, Constanța, Romania
| | - Victoria Birlutiu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Sibiu, Romania; County Clinical Emergency Hospital, Sibiu, Romania
| | - Irina M Dumitru
- Ovidius University of Constanța, Clinical Hospital of Infectious Diseases, Academy of Romanian Scientists, Bucharest, Romania
| | - Ruxandra Moroti
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; National Institute for Infectious Diseases Matei Bals, Bucharest, Romania
| | - Aleksandra Barac
- Clinic for Infectious and Tropical Diseases, University Clinical Center of Serbia, Belgrade, Serbia; Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Goran Stevanovic
- Clinic for Infectious and Tropical Diseases, University Clinical Center of Serbia, Belgrade, Serbia; Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Vasilios Koulouras
- Intensive Care Unit, University Hospital of Ioannina, University of Ioannina, Ioannina, Greece
| | | | | | - Emmanuel Roilides
- Infectious Diseases Unit, Hippokration General Hospital, Thessaloniki, Greece
| | - Alemka Markotić
- Dr Fran Mihaljević University Hospital for Infectious Diseases, Zagreb, Croatia
| | - Ushma Galal
- Nuffield Department of Primary Care Health Sciences, Clinical Trials Unit, University of Oxford, Oxford, UK
| | - Emmanuelle Denis
- ERGO, Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | - Lynsey Goodwin
- NIHR Health Protection Research Unit for Emerging Zoonotic Infections, University of Liverpool, Liverpool, UK
| | - Lance Turtle
- NIHR Health Protection Research Unit for Emerging Zoonotic Infections, University of Liverpool, Liverpool, UK
| | - Simin Aysel Florescu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Dr Victor Babes Clinical Hospital of Infectious and Tropical Diseases, Bucharest, Romania
| | - Hamdi Ramadani
- Clinic of Infectious Diseases, University Clinical Center of Kosovo, Prishtina, Kosovo; Department of Infectious Diseases, University Clinical Centre, Pristina, Kosovo
| | - Herman Goossens
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Margareta Ieven
- Department of Medical Microbiology, University of Antwerp UIA, Antwerp, Belgium; Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Christian Drosten
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Peter W Horby
- ERGO, Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | - Reina S Sikkema
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
8
|
Chun HM, Osawe S, Adams-Dabban S, Favaloro J, Iriemenam NC, Dirlikov E, Martin D, Milligan K, Abutu A, Okunoye O, Okoli M, Akanbi O, Akinmulero O, Okonkwo R, Oyedele O, Greby S, Abimiku A, Okoye MIJ, Shiraishi RW. SARS-CoV-2 serologic surveillance among people living with HIV in Nigeria, April 2022 to January 2023. Int J Infect Dis 2025; 151:107309. [PMID: 39579925 DOI: 10.1016/j.ijid.2024.107309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/18/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024] Open
Abstract
OBJECTIVES Evidence indicates that people living with HIV (PLHIV) are more impacted by COVID-19. The burden of SARS-CoV-2 infection among PLHIV is unknown in Nigeria. METHODS We conducted repeated cross-sectional SARS-CoV-2 serosurveys in 14 states and the Federal Capital Territory in Nigeria among PLHIV who had an HIV viral load (VL) test during April 2022 to January 2023. Evidence of SARS-CoV-2 immunoglobulin G (IgG) antibodies was assessed using a multiplex bead assay to measure IgG to spike (S), receptor binding domain (RBD), and nucleocapsid (N) proteins to identify potential infection and/or vaccination status. RESULTS Between April 2022 and January 2023, 47,614 remnant VL samples were included and tested for SARS-CoV-2 antibodies. Seroprevalence of SARS-CoV-2 infection, defined as IgG antibodies to spike and RBD591 [S+] and nucleocapsid [N+], (S+N+), ranged between 21.1% (95% confidence intervals [CI]: 11.4-31.8) in Ekiti State in January 2023 to 71.4% (95% CI 71.9-81.9) in Gombe State in November 2022, with overall steady trends within and between states over time, across age and sex. CONCLUSION High rates of SARS-CoV-2 antibody seroprevalence among PLHIV in Nigeria were observed. This underscores the need to understand the association between HIV and SARS-CoV-2 to inform strategies to reduce the threat posed by COVID-19.
Collapse
Affiliation(s)
- Helen M Chun
- Division of Global HIV & Tuberculosis, Global Health Center, US Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
| | - Sophia Osawe
- Institute of Human Virology, Abuja, Federal Capital Territory, Nigeria
| | | | - Jennifer Favaloro
- Division of Global HIV & Tuberculosis, Global Health Center, US Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Nnaemeka C Iriemenam
- Division of Global HIV & Tuberculosis, Global Health Center, US Centers for Disease Control and Prevention, Abuja, Federal Capital Territory, Nigeria
| | - Emilio Dirlikov
- Division of Global HIV & Tuberculosis, Global Health Center, US Centers for Disease Control and Prevention Mozambique, Maputo, Mozambique
| | - Diana Martin
- Division of Parasitic Diseases and Malaria, National Center for Emerging and Zoonotic Infectious Diseases, US Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Kyle Milligan
- Division of Global HIV & Tuberculosis, Global Health Center, US Centers for Disease Control and Prevention, Atlanta, Georgia, USA; Peraton Inc., Herndon, Virginia, USA
| | - Andrew Abutu
- Division of Global HIV & Tuberculosis, Global Health Center, US Centers for Disease Control and Prevention, Abuja, Federal Capital Territory, Nigeria
| | - Olumide Okunoye
- Division of Global HIV & Tuberculosis, Global Health Center, US Centers for Disease Control and Prevention, Abuja, Federal Capital Territory, Nigeria
| | - Mary Okoli
- Nigeria Centre for Disease Control and Prevention, Abuja, Federal Capital Territory, Nigeria
| | - Olusola Akanbi
- Nigeria Centre for Disease Control and Prevention, Abuja, Federal Capital Territory, Nigeria
| | | | - Rita Okonkwo
- Institute of Human Virology, Abuja, Federal Capital Territory, Nigeria
| | - Oyewole Oyedele
- Institute of Human Virology, Abuja, Federal Capital Territory, Nigeria
| | - Stacie Greby
- Division of Global HIV & Tuberculosis, Global Health Center, US Centers for Disease Control and Prevention, Abuja, Federal Capital Territory, Nigeria
| | - Alash'le Abimiku
- Institute of Human Virology, Abuja, Federal Capital Territory, Nigeria
| | - McPaul I J Okoye
- Division of Global HIV & Tuberculosis, Global Health Center, US Centers for Disease Control and Prevention, Abuja, Federal Capital Territory, Nigeria
| | - Ray W Shiraishi
- Division of Global HIV & Tuberculosis, Global Health Center, US Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
9
|
Lehmann PV, Karulin AY, Becza N, Yao L, Liu Z, Chepke J, Maul-Pavicic A, Wolf C, Köppert S, Valente AV, Gorbachev AV, Tary-Lehmann M, Kirchenbaum GA. Theoretical and practical considerations for validating antigen-specific B cell ImmunoSpot assays. J Immunol Methods 2025; 537:113817. [PMID: 39864733 DOI: 10.1016/j.jim.2025.113817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 10/17/2024] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
Owing to their ability to reliably detect even very rare antigen-specific B cells in cellular isolates such as peripheral blood mononuclear cells (PBMC), and doing so robustly in a high throughput-compatible manner, B cell ELISPOT/FluoroSpot (collectively "B cell ImmunoSpot") tests have become increasingly attractive for immune monitoring in regulated settings. Presently, there are no guidelines for the qualification and validation of B cell ImmunoSpot assay results. Here, we propose such guidelines, building on the experience acquired from T cell ImmunoSpot testing in an environment adhering to the requirements of regulatory bodies yet taking the unique features of B cell assays into account. A streamlined protocol is proposed that permits the performance of all tests needed for the formal validation of an antigen-specific B cell ImmunoSpot assay in only three experiments, utilizing 2.2 × 107 PBMC per donor. Subsequently, utilizing only 1-2 × 106 PBMC per sample (obtainable from 1 to 2 mL of blood), a validated multiplexed assay enables accurate quantification of the frequency of antigen-specific memory B cell-derived blasts secreting IgM, IgG, IgA or IgE antibodies. Collectively, such multiplexed B cell ImmunoSpot assays offer immense value for B cell immune monitoring programs due to their ease of implementation, scalability, applicability to essentially any antigenic system, economy of PBMC utilization, and last but not least, the high content information gained.
Collapse
Affiliation(s)
- Paul V Lehmann
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Alexey Y Karulin
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Noémi Becza
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Lingling Yao
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Zhigang Liu
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Jack Chepke
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Andrea Maul-Pavicic
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Carla Wolf
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Sebastian Köppert
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Alexis V Valente
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Anton V Gorbachev
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Magdalena Tary-Lehmann
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA
| | - Greg A Kirchenbaum
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| |
Collapse
|
10
|
Masuda K, Iketani S, Liu L, Huang J, Qiao Y, Shah J, McNairy ML, Groso C, Ricupero C, Loffredo LF, Wang Q, Purpura L, Coelho-dos-Reis JGA, Sheng Z, Yin MT, Tsuji M. Distinct CD8 + T-cell types Associated with COVID-19 Severity in Unvaccinated HLA-A2 + Patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.12.632164. [PMID: 39868279 PMCID: PMC11761488 DOI: 10.1101/2025.01.12.632164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Although emerging data have revealed the critical role of memory CD8+ T cells in preventing and controlling SARS-CoV-2 infection, virus-specific CD8+ T-cell responses against SARS-CoV-2 and its memory and innate-like subsets in unvaccinated COVID-19 patients with various disease manifestations in an HLA-restricted fashion remain to be understood. Here, we show the strong association of protective cellular immunity with mild COVID-19 and unique cell types against SARS-CoV-2 virus in an HLA-A2 restricted manner. ELISpot assays reveal that SARS-CoV-2-specific CD8+ T-cell responses in mild COVID-19 patients are significantly higher than in severe patients, whereas neutralizing antibody responses against SARS-CoV-2 virus significantly correlate with disease severity. Single-cell analyses of HLA-A2-restricted CD8+ T cells, which recognize highly conserved immunodominant SARS-CoV-2-specific epitopes, demonstrate divergent profiles in unvaccinated patients with mild versus severe disease. CD8+ T-cell types including cytotoxic KLRB1 + CD8αα cells with innate-like T-cell signatures, IFNG hi ID3 hi memory cells and IL7R + proliferative stem cell-like memory cells are preferentially observed in mild COVID-19, whereas distinct terminally-differentiated T-cell subsets are predominantly detected in severe COVID-19: highly activated FASL hi T-cell subsets and early-terminated or dysfunctional IL4R + GATA3 + stem cell-like memory T-cell subset. In conclusion, our findings suggest that unique and contrasting SARS-CoV-2-specific CD8+ T-cell profiles may dictate COVID-19 severity.
Collapse
Affiliation(s)
- Kazuya Masuda
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Sho Iketani
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Lihong Liu
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jing Huang
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Yujie Qiao
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jayesh Shah
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Meredith L. McNairy
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Christine Groso
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Christopher Ricupero
- Center for Dental & Craniofacial Regeneration, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Lucas F. Loffredo
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Qian Wang
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Lawrence Purpura
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | | | - Zizhang Sheng
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Michael T Yin
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Lead contact
| |
Collapse
|
11
|
Bailey J, Lavelle B, Miller J, Jimenez M, Lim PH, Orban ZS, Clark JR, Tomar R, Ludwig A, Ali ST, Lank GK, Zielinski A, Mylvaganam R, Kalhan R, El Muayed M, Mutharasan RK, Liotta EM, Sznajder JI, Davidson C, Koralnik IJ, Sala MA. Multidisciplinary Center Care for Long COVID Syndrome-A Retrospective Cohort Study. Am J Med 2025; 138:108-120. [PMID: 37220832 PMCID: PMC10200714 DOI: 10.1016/j.amjmed.2023.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/17/2023] [Accepted: 05/17/2023] [Indexed: 05/25/2023]
Abstract
BACKGROUND Persistent multi-organ symptoms after coronavirus disease 2019 (COVID-19) have been termed "long COVID" or "post-acute sequelae of SARS-CoV-2 infection." The complexity of these clinical manifestations posed challenges early in the pandemic as different ambulatory models formed out of necessity to manage the influx of patients. Little is known about the characteristics and outcomes of patients seeking care at multidisciplinary post-COVID centers. METHODS We performed a retrospective cohort study of patients evaluated at our multidisciplinary comprehensive COVID-19 center in Chicago, Ill, between May 2020 and February 2022. We analyzed specialty clinic utilization and clinical test results according to severity of acute COVID-19. RESULTS We evaluated 1802 patients a median of 8 months from acute COVID-19 onset, including 350 post-hospitalization and 1452 non-hospitalized patients. Patients were seen in 2361 initial visits in 12 specialty clinics, with 1151 (48.8%) in neurology, 591 (25%) in pulmonology, and 284 (12%) in cardiology. Among the patients tested, 742/916 (81%) reported decreased quality of life, 284/553 (51%) had cognitive impairment, 195/434 (44.9%) had alteration of lung function, 249/299 (83.3%) had abnormal computed tomography chest scans, and 14/116 (12.1%) had elevated heart rate on rhythm monitoring. Frequency of cognitive impairment and pulmonary dysfunction was associated with severity of acute COVID-19. Non-hospitalized patients with positive SARS-CoV-2 testing had findings similar to those with negative or no test results. CONCLUSIONS The experience at our multidisciplinary comprehensive COVID-19 center shows common utilization of multiple specialists by long COVID patients, who harbor frequent neurologic, pulmonary, and cardiologic abnormalities. Differences in post-hospitalization and non-hospitalized groups suggest distinct pathogenic mechanisms of long COVID in these populations.
Collapse
Affiliation(s)
- Joseph Bailey
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill.
| | - Bianca Lavelle
- McGaw Medical Center, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Janet Miller
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Millenia Jimenez
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Patrick H Lim
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Zachary S Orban
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Jeffrey R Clark
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Ria Tomar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Amy Ludwig
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Sareen T Ali
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Grace K Lank
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Allison Zielinski
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Ruben Mylvaganam
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Ravi Kalhan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Malek El Muayed
- Division of Endocrinology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - R Kannan Mutharasan
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Eric M Liotta
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Jacob I Sznajder
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Charles Davidson
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Igor J Koralnik
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Marc A Sala
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| |
Collapse
|
12
|
Pickering S, Wilson H, Bravo E, Perera MR, Seow J, Graham C, Almeida N, Fotopoulos L, Williams T, Moitra A, Winstone H, Nissen TAD, Galão RP, Snell LB, Doores KJ, Malim MH, Neil SJD. Antibodies to the RBD of SARS-CoV-2 spike mediate productive infection of primary human macrophages. Nat Commun 2024; 15:10764. [PMID: 39737903 PMCID: PMC11686093 DOI: 10.1038/s41467-024-54458-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/07/2024] [Indexed: 01/01/2025] Open
Abstract
The role of myeloid cells in the pathogenesis of SARS-CoV-2 is well established, in particular as drivers of cytokine production and systemic inflammation characteristic of severe COVID-19. However, the potential for myeloid cells to act as bona fide targets of productive SARS-CoV-2 infection, and the specifics of entry, remain unclear. Using a panel of anti-SARS-CoV-2 monoclonal antibodies (mAbs) we performed a detailed assessment of antibody-mediated infection of monocytes/macrophages. mAbs with the most consistent potential to mediate infection were those targeting a conserved region of the receptor binding domain (RBD; group 1/class 4). Infection was closely related to the neutralising concentration of the mAbs, with peak infection occurring below the IC50, while pre-treating cells with remdesivir or FcγRI-blocking antibodies inhibited infection. Studies performed in primary macrophages demonstrated high-level and productive infection, with infected macrophages appearing multinucleated and syncytial. Infection was not seen in the absence of antibody with the same quantity of virus. Addition of ruxolitinib significantly increased infection, indicating restraint of infection through innate immune mechanisms rather than entry. High-level production of pro-inflammatory cytokines directly correlated with macrophage infection levels. We hypothesise that infection via antibody-FcR interactions could contribute to pathogenesis in primary infection, systemic virus spread or persistent infection.
Collapse
MESH Headings
- Humans
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
- Macrophages/immunology
- Macrophages/virology
- Macrophages/metabolism
- SARS-CoV-2/immunology
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- COVID-19/immunology
- COVID-19/virology
- Antibodies, Viral/immunology
- Nitriles/pharmacology
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/pharmacology
- Pyrimidines/pharmacology
- Pyrazoles/pharmacology
- Alanine/analogs & derivatives
- Alanine/pharmacology
- Receptors, IgG/metabolism
- Receptors, IgG/immunology
- Adenosine Monophosphate/analogs & derivatives
- Adenosine Monophosphate/pharmacology
- Protein Domains
- Cells, Cultured
- Virus Internalization/drug effects
Collapse
Affiliation(s)
- Suzanne Pickering
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK.
| | - Harry Wilson
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Enrico Bravo
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Marianne R Perera
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Jeffrey Seow
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Carl Graham
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Nathalia Almeida
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Lazaros Fotopoulos
- The Stem Cell Hotel, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London, SE1 9RT, UK
- Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London, SE1 9RT, UK
| | - Thomas Williams
- The Stem Cell Hotel, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London, SE1 9RT, UK
- Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London, SE1 9RT, UK
| | - Atlanta Moitra
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Helena Winstone
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Tinne A D Nissen
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
| | - Rui Pedro Galão
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Luke B Snell
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy's and St Thomas' NHS Foundation Trust, London, SE1 7EH, UK
| | - Katie J Doores
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Michael H Malim
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Stuart J D Neil
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| |
Collapse
|
13
|
Yamamoto M, Sakurai K, Takatani R, Hisada A, Mori C. Three-year seroprevalence of SARS-CoV-2 nucleocapsid protein antibody among children, parental awareness, and contributors of infection: a single-school cohort study in Chiba, Japan. J Epidemiol 2024; 35:278-286. [PMID: 39710421 PMCID: PMC12066190 DOI: 10.2188/jea.je20240284] [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: 03/21/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) in children is often asymptomatic, posing challenges in detecting infections. Additionally, factors contributing to infection remain poorly understood. This study aimed to investigate trends in anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleocapsid antibody seroprevalence, the relationship between seroprevalence and parental perception of child infection, and factors related to COVID-19 in children. METHODS In December 2020, 355 children aged 6-12 years in one elementary school were enrolled in the study. The anti-SARS-CoV-2 nucleocapsid antibody seroprevalence was assessed, and questionnaires were administered annually for three years. Parents' perceptions of infection and factors contributing to infection were examined. RESULTS The seroprevalence was 0.6%, 2.2%, and 60.9% in the first, second, and third years, respectively. The third-year seroprevalence among children reported as 'infected,' 'not tested but had symptoms,' and 'not infected' by parents was 97.3%, 83.3%, and 35.7%, respectively. Increased odds of seropositivity at the third-year measurement were observed in lower grades (adjusted odds ratio [aOR]=2.79 compared with higher grades) and in children more likely to play with others (aOR=3.97 for 'somewhat' and 2.84 for 'often,' compared with 'rarely'). No significant associations with seropositivity were found for sex, siblings, body mass index, serum 25-OH vitamin D3 concentration, or sleep duration. CONCLUSION The Omicron variant outbreak from the end of 2021 led to a sharp increase in seroprevalence among children, with many unaware of their infection. Frequent play with others may facilitate transmission in children. These data provide useful information for developing countermeasures against COVID-19 and other future pandemics.
Collapse
Affiliation(s)
- Midori Yamamoto
- Department of Sustainable Health Science, Center for Preventive Medical Sciences, Chiba University, Chiba, Japan
| | - Kenichi Sakurai
- Department of Nutrition and Metabolic Medicine, Center for Preventive Medical Sciences, Chiba University, Chiba, Japan
| | - Rieko Takatani
- Department of Clinical Medicine, Faculty of Education, Chiba University, Chiba, Japan
| | - Aya Hisada
- Department of Sustainable Health Science, Center for Preventive Medical Sciences, Chiba University, Chiba, Japan
| | - Chisato Mori
- Department of Sustainable Health Science, Center for Preventive Medical Sciences, Chiba University, Chiba, Japan
- Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
14
|
Harris AW, Kurtovic L, Nogueira J, Bouzas I, Opi DH, Wines BD, Lee WS, Hogarth PM, Poumbourios P, Drummer HE, Valim C, Porto LC, Beeson JG. Induction of Fc-dependent functional antibodies against different variants of SARS-CoV-2 varies by vaccine type and prior infection. COMMUNICATIONS MEDICINE 2024; 4:273. [PMID: 39702507 DOI: 10.1038/s43856-024-00686-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 11/21/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND SARS-CoV-2 transmission and COVID-19 disease severity is influenced by immunity from natural infection and/or vaccination. Population-level immunity is complicated by the emergence of viral variants. Antibody Fc-dependent effector functions are as important mediators in immunity. However, their induction in populations with diverse infection and/or vaccination histories and against variants remains poorly defined. METHODS We evaluated Fc-dependent functional antibodies following vaccination with two widely used vaccines, AstraZeneca (AZ) and Sinovac (SV), including antibody binding of Fcγ-receptors and complement-fixation in vaccinated Brazilian adults (n = 222), some of who were previously infected with SARS-CoV-2, as well as adults with natural infection only (n = 200). IgG, IgM, IgA, and IgG subclasses were also quantified. RESULTS AZ induces greater Fcγ-receptor-binding (types I, IIa, and IIIa/b) antibodies than SV or natural infection. Previously infected individuals have significantly greater vaccine-induced responses compared to naïve counterparts. Fcγ-receptor-binding is highest among AZ vaccinated individuals with a prior infection, for all receptor types, and substantial complement-fixing activity is only seen among this group. SV induces higher IgM than AZ, but this does not drive better complement-fixing activity. Some SV responses are associated with subject age, whereas AZ responses are not. Importantly, functional antibody responses are well retained against the Omicron BA.1 S protein, being best retained for Fcγ-receptor-1 binding, and are higher for AZ than SV. CONCLUSIONS Hybrid immunity, from combined natural exposure and vaccination, generates strong Fc-mediated antibody functions which may contribute to immunity against evolving SARS-CoV-2 variants. Understanding determinants of Fc-mediated functions may enable future vaccines with greater efficacy against different variants.
Collapse
Affiliation(s)
- Alexander W Harris
- Burnet Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne, Australia
| | - Liriye Kurtovic
- Burnet Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne, Australia
| | - Jeane Nogueira
- Immunogenic and Histocompatibility Laboratory, Technologic Core for Tissue repair and Histocompatibility, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Isabel Bouzas
- Health Research Support Facility Center (CAPCS), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - D Herbert Opi
- Burnet Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne, Australia
- Departments of Medicine, The University of Melbourne, Melbourne, Australia
| | - Bruce D Wines
- Burnet Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne, Australia
| | - Wen Shi Lee
- Department of Microbiology and Immunology at The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | | | - Pantelis Poumbourios
- Burnet Institute, Melbourne, Australia
- Department of Microbiology, Monash University, Melbourne, Australia
| | - Heidi E Drummer
- Burnet Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne, Australia
- Department of Microbiology and Immunology at The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Clarissa Valim
- Boston University School of Public Health, Boston University, Boston, USA
| | - Luís Cristóvão Porto
- Immunogenic and Histocompatibility Laboratory, Technologic Core for Tissue repair and Histocompatibility, Rio de Janeiro State University, Rio de Janeiro, Brazil
- Health Research Support Facility Center (CAPCS), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - James G Beeson
- Burnet Institute, Melbourne, Australia.
- Department of Immunology, Monash University, Melbourne, Australia.
- Departments of Medicine, The University of Melbourne, Melbourne, Australia.
- Department of Microbiology and Immunology at The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
15
|
Kusunoki H. Current Status and Significance of Additional Vaccination with COVID-19 Vaccine in Japan-Considerations from Antibody Levels from Hybrid Immunity and Public Perceptions. Vaccines (Basel) 2024; 12:1413. [PMID: 39772074 PMCID: PMC11680318 DOI: 10.3390/vaccines12121413] [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: 11/24/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
This report examines the evolving role of coronavirus disease 2019 (COVID-19) vaccination in Japan, especially in light of the reduced public concern following the reclassification of COVID-19 as a Category 5 infectious disease in May 2023. With over half the population estimated to have hybrid immunity from prior infections and vaccinations, this report evaluated the necessity and frequency of additional booster doses. Despite strong recommendations from Japanese medical societies to continue vaccination, public skepticism remains owing to financial burdens, adverse reactions, and the perceived limited benefits of frequent boosters. Studies on antibody responses have revealed that individuals with hybrid immunity maintain robust protection with significantly elevated antibody titers that persist over extended periods. Case studies have indicated durable immunity among individuals who have both been vaccinated and experienced breakthrough infections, raising questions about the need for uniform booster policies. This report also discusses the newly approved replicon-type (self-amplifying) vaccines currently available only in Japan, which have generated public and professional debates regarding their efficacy and safety. A more personalized approach to vaccination that takes into account the antibody titers, prior infection history, and individual choices is recommended. Finally, this report underscores the importance of aligning vaccination policies with scientific evidence and public sentiment to optimize COVID-19 countermeasures in Japan.
Collapse
Affiliation(s)
- Hiroshi Kusunoki
- Department of Internal Medicine, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata 573-1121, Osaka, Japan
| |
Collapse
|
16
|
Odle A, Kar M, Verma AK, Sariol A, Meyerholz DK, Suthar MS, Wong LYR, Perlman S. Tissue-resident memory T cells contribute to protection against heterologous SARS-CoV-2 challenge. JCI Insight 2024; 9:e184074. [PMID: 39405115 PMCID: PMC11623939 DOI: 10.1172/jci.insight.184074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
New vaccine formulations are based on circulating strains of virus, which have tended to evolve to more readily transmit human to human and to evade the neutralizing antibody response. An assumption of this approach is that ancestral strains of virus will not recur. Recurrence of these strains could be a problem for individuals not previously exposed to ancestral spike protein. Here, we addressed this by infecting mice with recent SARS-CoV-2 variants and then challenging them with a highly pathogenic mouse-adapted virus closely related to the ancestral Wuhan-1 strain (SARS2-N501YMA30). We found that challenged mice were protected from severe disease, despite having low or no neutralizing antibodies against SARS2-N501YMA30. T cell depletion from previously infected mice did not diminish infection against clinical disease, although it resulted in delayed virus clearance in the nasal turbinate and, in some cases, in the lungs. Levels of tissue-resident memory T cells were significantly elevated in the nasal turbinate of previously infected mice compared with that of naive mice. However, this phenotype was not seen in lung tissues. Together, these results indicate that the immune response to newly circulating variants afforded protection against reinfection with the ancestral virus that was in part T cell based.
Collapse
Affiliation(s)
- Abby Odle
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Meenakshi Kar
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Vaccine Center, Atlanta, Georgia, USA
| | - Abhishek K. Verma
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Alan Sariol
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Mehul S. Suthar
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Vaccine Center, Atlanta, Georgia, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lok-Yin Roy Wong
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
- Department of Microbiology, Biochemistry and Molecular Genetics and
- Center for Virus-Host-Innate Immunity, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
17
|
Kimotho J, Sein Y, Sayed S, Shah R, Mwai K, Saleh M, Wanjiku P, Mwacharo J, Nyagwange J, Karanja H, Kutima B, Gitonga JN, Mugo D, Karanu A, Moranga L, Oluoch V, Shah J, Mutiso J, Mburu A, Nneka Z, Betti P, Usyu Mutinda W, Issak Abdi A, Bejon P, Isabella Ochola-Oyier L, M.Warimwe G, Nduati EW, M. Ndungu F. Kinetics of naturally induced binding and neutralising anti-SARS-CoV-2 antibody levels and potencies among SARS-CoV-2 infected Kenyans with diverse grades of COVID-19 severity: an observational study. Wellcome Open Res 2024; 8:350. [PMID: 39640868 PMCID: PMC11617823 DOI: 10.12688/wellcomeopenres.19414.2] [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] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
Background Given the low levels of coronavirus disease 2019 (COVID-19) vaccine coverage in sub-Saharan Africa (sSA), despite high levels of natural severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) exposures, strategies for extending the breadth and longevity of naturally acquired immunity are warranted. Designing such strategies will require a good understanding of naturally acquired immunity. Methods We measured whole-spike immunoglobulin G (IgG) and spike-receptor binding domain (RBD) total immunoglobulins (Igs) on 585 plasma samples collected longitudinally over five successive time points within six months of COVID-19 diagnosis in 309 COVID-19 patients. We measured antibody-neutralising potency against the wild-type (Wuhan) SARS-CoV-2 pseudovirus in a subset of 51 patients over three successive time points. Binding and neutralising antibody levels and potencies were then tested for correlations with COVID-19 severities. Results Rates of seroconversion increased from day 0 (day of PCR testing) to day 180 (six months) (63.6% to 100 %) and (69.3 % to 97%) for anti-spike-IgG and anti-spike-RBD binding Igs, respectively. Levels of these binding antibodies peaked at day 28 (p<0.01) and were subsequently maintained for six months without significant decay (p>0.99). Similarly, antibody-neutralising potencies peaked at day 28 (p<0.01) but declined by three-fold, six months after COVID-19 diagnosis (p<0.01). Binding antibody levels were highly correlated with neutralising antibody potencies at all the time points analysed (r>0.60, p<0.01). Levels and potencies of binding and neutralising antibodies increased with disease severity. Conclusions Most COVID-19 patients generated SARS-CoV-2 specific binding antibodies that remained stable in the first six months of infection. However, the respective neutralising antibodies decayed three-fold by month-six of COVID-19 diagnosis suggesting that they are short-lived, consistent with what has been observed elsewhere in the world. Thus, regular vaccination boosters are required to sustain the high levels of anti-SARS-CoV-2 naturally acquired neutralising antibody potencies in our population.
Collapse
Affiliation(s)
- John Kimotho
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Pwani University, KILIFI, 230-80108, Kenya
| | - Yiakon Sein
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Shahin Sayed
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Reena Shah
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Kennedy Mwai
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Mansoor Saleh
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Perpetual Wanjiku
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Jedidah Mwacharo
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - James Nyagwange
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Henry Karanja
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Bernadette Kutima
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - John N. Gitonga
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Daisy Mugo
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Ann Karanu
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Linda Moranga
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
| | - Viviane Oluoch
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Jasmit Shah
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Julius Mutiso
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Alfred Mburu
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Zaitun Nneka
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | - Peter Betti
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
| | | | - Abdirahman Issak Abdi
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Pwani University, KILIFI, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Philip Bejon
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Lynette Isabella Ochola-Oyier
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - George M.Warimwe
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Eunice W. Nduati
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Pwani University, KILIFI, 230-80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Francis M. Ndungu
- KEMRI-Wellcome Trust Research Programme, KILIFI, Coast, 230-80108, Kenya
- Pwani University, KILIFI, 230-80108, Kenya
- Aga Khan University Hospital, 3rd Parklands Avenue, Nairobi, 30270 - 00100, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Division of Infectious Diseases, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
18
|
Yan Q, Zhang Y, Hou R, Pan W, Liang H, Gao X, Deng W, Huang X, Qu L, Tang C, He P, Liu B, Wang Q, Zhao X, Lin Z, Chen Z, Li P, Han J, Xiong X, Zhao J, Li S, Niu X, Chen L. Deep immunoglobulin repertoire sequencing depicts a comprehensive atlas of spike-specific antibody lineages shared among COVID-19 convalescents. Emerg Microbes Infect 2024; 13:2290841. [PMID: 38044868 PMCID: PMC10810631 DOI: 10.1080/22221751.2023.2290841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/29/2023] [Indexed: 12/05/2023]
Abstract
Neutralizing antibodies are a key component in protective humoral immunity against SARS-CoV-2. Currently, available technologies cannot track epitope-specific antibodies in global antibody repertoires. Thus, the comprehensive repertoire of spike-specific neutralizing antibodies elicited by SARS-CoV-2 infection is not fully understood. We therefore combined high-throughput immunoglobulin heavy chain (IgH) repertoire sequencing, and structural and bioinformatics analysis to establish an antibodyomics pipeline, which enables tracking spike-specific antibody lineages that target certain neutralizing epitopes. We mapped the neutralizing epitopes on the spike and determined the epitope-preferential antibody lineages. This analysis also revealed numerous overlaps between immunodominant neutralizing antibody-binding sites and mutation hotspots on spikes as observed so far in SARS-CoV-2 variants. By clustering 2677 spike-specific antibodies with 360 million IgH sequences that we sequenced, a total of 329 shared spike-specific antibody clonotypes were identified from 33 COVID-19 convalescents and 24 SARS-CoV-2-naïve individuals. Epitope mapping showed that the shared antibody responses target not only neutralizing epitopes on RBD and NTD but also non-neutralizing epitopes on S2. The immunodominance of neutralizing antibody response is determined by the occurrence of specific precursors in human naïve B-cell repertoires. We identified that only 28 out of the 329 shared spike-specific antibody clonotypes persisted for at least 12 months. Among them, long-lived IGHV3-53 antibodies are likely to evolve cross-reactivity to Omicron variants through accumulating somatic hypermutations. Altogether, we created a comprehensive atlas of spike-targeting antibody lineages in COVID-19 convalescents and antibody precursors in human naïve B cell repertoires, providing a valuable reference for future vaccine design and evaluation.
Collapse
Affiliation(s)
- Qihong Yan
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Yudi Zhang
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
- University of Chinese Academy of Science, Beijing, People’s Republic of China
| | - Ruitian Hou
- Guangzhou Institute of Infectious Disease, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Wenjing Pan
- Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
- Nanjing ARP Biotechnology Co., Ltd, Nanjing, People’s Republic of China
| | - Huan Liang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Xijie Gao
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Weiqi Deng
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
- University of Chinese Academy of Science, Beijing, People’s Republic of China
| | - Xiaohan Huang
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Linbing Qu
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
| | - Congli Tang
- Nanjing ARP Biotechnology Co., Ltd, Nanjing, People’s Republic of China
| | - Ping He
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
- Guangzhou National Laboratory, Guangzhou, People’s Republic of China
| | - Banghui Liu
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
| | - Qian Wang
- Guangzhou National Laboratory, Guangzhou, People’s Republic of China
| | - Xinwei Zhao
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
- Guangzhou National Laboratory, Guangzhou, People’s Republic of China
| | - Zihan Lin
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
- University of Chinese Academy of Science, Beijing, People’s Republic of China
| | - Zhaoming Chen
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
| | - Pingchao Li
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
| | - Jian Han
- iRepertoire Inc., Huntsville, AL, USA
| | - Xiaoli Xiong
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
- Guangzhou National Laboratory, Guangzhou, People’s Republic of China
| | - Song Li
- Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Xuefeng Niu
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
- Guangzhou Institute of Infectious Disease, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
- Guangzhou National Laboratory, Guangzhou, People’s Republic of China
| |
Collapse
|
19
|
Krishna B, Metaxaki M, Perera M, Wills M, Sithole N. Comparison of different T cell assays for the retrospective determination of SARS-CoV-2 infection. J Gen Virol 2024; 105. [PMID: 39704047 DOI: 10.1099/jgv.0.002055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
It is important to be able to retrospectively determine severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections with high accuracy, both for post-coronavirus disease 2019 (COVID-19) epidemiological studies, and to distinguish between Long COVID and other multi-syndromic diseases that have overlapping symptoms. Although serum antibody levels can be measured to retrospectively diagnose SARS-CoV-2 infections, peptide stimulation of memory T cell responses is a more sensitive approach. This is because robust memory T cells are generated after SARS-CoV-2 infection and persist even after antibodies wane below detectability thresholds. In this study, we compare T cell responses using FluoroSpot-based methods and overnight stimulation of whole blood with SARS-CoV-2 peptides followed by an ELISA. Both approaches have comparable sensitivity and specificity but require different equipment and samples to be used. Furthermore, the elimination of peptides that cross-react with other coronaviruses increases the assay specificity but trades off some sensitivity. Finally, this approach can be used on archival, cryopreserved PBMCs. This work shows comparative advantages for several methods to measure SARS-CoV-2 T cell responses that could be utilized by any laboratory studying the effects of the coronavirus disease 2019 pandemic.
Collapse
Affiliation(s)
- Benjamin Krishna
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Marina Metaxaki
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Marianne Perera
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Mark Wills
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Nyarie Sithole
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| |
Collapse
|
20
|
Zhang H, Li B, Sun J, Zou L, Yi L, Lin H, Zhou P, Liang C, Zeng L, Zhuang X, Liu Z, Lu J, He J, Yuan R. Immune evasion after SARS-CoV-2 Omicron BA.5 and XBB.1.9 endemic observed from Guangdong Province, China from 2022 to 2023. Virol J 2024; 21:298. [PMID: 39568037 PMCID: PMC11577657 DOI: 10.1186/s12985-024-02573-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND From 2022 to 2023, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused by Omicron variants spread rapidly in Guangdong Province, resulting in over 80% of the population being infected. RESULTS To investigate the levels of neutralizing antibodies (NAbs) in individuals following the rapid pandemic and to evaluate the cross-protection against currently circulating variants of SARS-CoV-2 in China, neutralization assay and magnetic particle chemiluminescence method were used to test the 117 serum samples from individuals who had recovered 4 weeks post-infection. The results indicated that the levels of NAbs against prototype and Omicron variants BA.5 were significantly higher than those against Omicron variants BQ.1, XBB.1.1, XBB.1.9, XBB.1.16 and EG.5, regardless of whether the infection was primary or secondary. CONCLUSIONS The cross-protection provided by NAbs induced by prototype and Omicron BA.5 variants was limited when challenged by BQ.1, XBB.1.1, XBB.1.9, XBB.1.16 and EG.5 variants. This indicates that we should pay more attention to the risk of multiple infection from any novel Omicron variants that may emerge in the near future.
Collapse
Affiliation(s)
- Huan Zhang
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Baisheng Li
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Jiufeng Sun
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, 510632, China
- School of Public Health, Southern Medical University, Guangzhou, 510515, China
- School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510310, China
| | - Lirong Zou
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Lina Yi
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Huifang Lin
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Pingping Zhou
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Chumin Liang
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Lilian Zeng
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Xue Zhuang
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Zhe Liu
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Jing Lu
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Jianfeng He
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China
| | - Runyu Yuan
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, 511430, China.
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China.
| |
Collapse
|
21
|
Seow J, Jefferson GCE, Keegan MD, Yau Y, Snell LB, Doores KJ. Profiling serum immunodominance following SARS-CoV-2 primary and breakthrough infection reveals distinct variant-specific epitope usage and immune imprinting. PLoS Pathog 2024; 20:e1012724. [PMID: 39556615 DOI: 10.1371/journal.ppat.1012724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/03/2024] [Indexed: 11/20/2024] Open
Abstract
Over the course of the COVID-19 pandemic, variants have emerged with increased mutations and immune evasive capabilities. This has led to breakthrough infections (BTI) in vaccinated individuals, with a large proportion of the neutralizing antibody response targeting the receptor binding domain (RBD) of the SARS-CoV-2 Spike glycoprotein. Immune imprinting, where prior exposure of the immune system to an antigen can influence the response to subsequent exposures, and its role in a population with heterogenous exposure histories has important implications in future vaccine design. Here, we develop an accessible approach to map epitope immunodominance of the neutralizing antibody response in sera. By using a panel of mutant Spike proteins in a pseudotyped virus neutralization assay, we observed distinct epitope usage in convalescent donors infected during wave 1, or infected with the Delta, or BA.1 variants, highlighting the antigenic diversity of the variant Spikes. Analysis of longitudinal serum samples taken spanning 3 doses of COVID-19 vaccine and subsequent breakthrough infection, showed the influence of immune imprinting from the ancestral-based vaccine, where reactivation of existing B cells elicited by the vaccine resulted in the enrichment of the pre-existing epitope immunodominance. However, subtle shifts in epitope usage in sera were observed following BTI by Omicron sub-lineage variants. Antigenic distance of Spike, time after last exposure, and number of vaccine boosters may play a role in the persistence of imprinting from the vaccine. This study provides insight into RBD neutralizing epitope usage in individuals with varying exposure histories and has implications for design of future SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Jeffrey Seow
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - George C E Jefferson
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Michael D Keegan
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Yeuk Yau
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Luke B Snell
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Katie J Doores
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
22
|
Wang L, Xu R, Huang D, Peng P, Sun K, Hu J, Liu BZ, Fang L, Zhang L, Sun X, Gu F, Tang N, Huang AL, Lin X, Lan X. Identification of virus epitopes and reactive T-cell receptors from memory T cells without peptide synthesis. Commun Biol 2024; 7:1432. [PMID: 39496850 PMCID: PMC11535475 DOI: 10.1038/s42003-024-07048-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/10/2024] [Indexed: 11/06/2024] Open
Abstract
Identifying epitopes and their corresponding T-cell receptor (TCR) sequences is crucial in the face of rapidly mutating viruses. Peptide synthesis is often required to confirm the exact epitope sequences, which is time-consuming and expensive. In this study, we introduce a scalable workflow to identify the exact sequences of virus epitopes and reactive TCRs targeting the epitopes from memory T cells. Following the narrowing down of epitopes to specific regions via the tandem minigene (TMG) system, our workflow incorporates the utilization of peptide-major histocompatibility complex-displaying yeasts (pMHC-displaying yeasts) to rapidly screen immunogenic epitopes' precise sequences, obviating the necessity for the chemical synthesis of peptides. Focusing on SARS-CoV-2, we identify the precise sequences of reactive TCRs, targeting conserved epitopes across the Coronaviridae family, from the blood of COVID-19-recovered individuals over 8 months. Notably, we reveal that at least 75% (6/8) of the tested donors harbor T cells targeting a shared epitope, KTFPPTEPK, derived from the N protein. Furthermore, several identified TCRs exhibit cross-reactivity to mutant epitopes, suggesting a potential mechanism for sustained T-cell responses against emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Lihui Wang
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Runda Xu
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Daosheng Huang
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Pai Peng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Keyong Sun
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Jie Hu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Bei-Zhong Liu
- Yong-Chuan Hospital of Chongqing Medical University, Chongqing, China
| | - Liang Fang
- Yong-Chuan Hospital of Chongqing Medical University, Chongqing, China
| | - Liwen Zhang
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Xin Sun
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Fei Gu
- Alibaba Group, 311121, Hangzhou, China.
| | - Ni Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
| | - Ai-Long Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
| | - Xin Lin
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China.
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China.
| | - Xun Lan
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China.
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China.
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
23
|
Chen Y, Zhang X, Zhou M, Wu P, Yan J, Sun C, Zhang Y, Zheng X. Antibody response to SARS-CoV-2 natural and breakthrough infection in patients undergoing maintenance hemodialysis: A prospective cohort study over 4 months. Heliyon 2024; 10:e38545. [PMID: 39397907 PMCID: PMC11470422 DOI: 10.1016/j.heliyon.2024.e38545] [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: 11/22/2023] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024] Open
Abstract
Background Patients undergoing maintenance hemodialysis (MHD) are susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. However, the antibody response of these patients to natural and breakthrough infections remains poorly understood. Methods Between January 15, 2023, and February 15, 2023, a total of 53 patients undergoing MHD and diagnosed with SARS-CoV-2 infection at The Affiliated Hospital of Hangzhou Normal University were enrolled. They were categorized into the natural (n = 40) and breakthrough infection groups (n = 13) based on their vaccination status before infection. Comprehensive data, including basic clinical information, vaccination status, and routine post-infection blood parameters, were collected from all participants. Blood specimens were drawn monthly after infection, and SARS-CoV-2 receptor-binding domain (RBD) immunoglobulin (Ig) G and IgM tests were conducted. The study included continuous follow-up over 4 months. Results During the acute phase of infection, the SARS-CoV-2 RBD IgM positivity was 5 % in patients undergoing MHD who were naturally infected and 15.4 % in those with breakthrough infections. Four months after infection, SARS-CoV-2 RBD IgG positivity in patients with natural and breakthrough infection was 77.5 % and 100 %, respectively. Patients undergoing MHD with breakthrough infection exhibited higher SARS-CoV-2 RBD IgG titers (751.21 signal-to cutoff ratio, S/CO [interquartile range, IQR, 30.54-1173.63]) than those with natural infections (3.43 S/CO [IQR, 1.12-15.6]) (p < 0.0001). No significant differences were observed in SARS-CoV-2 RBD IgG between males and females or among those aged <70 and ≥70 years. Patients who received three doses of the inactivated vaccine produced significantly higher SARS-CoV-2 RBD IgG levels after infection than those who received one or two doses; these differences were significant. Conclusions Although patients undergoing MHD exhibit a low rate of SARS-CoV-2 RBD IgM positivity following infection, those vaccinated with inactivated vaccines can generate elevated SARS-CoV-2 RBD IgG levels, particularly those who receive three doses.
Collapse
Affiliation(s)
- Yingji Chen
- Department of Nephrology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Xiaming Zhang
- Department of Nephrology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Mi Zhou
- Department of Nephrology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Ping Wu
- Department of Nephrology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Juzhen Yan
- Department of Nephrology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Chen Sun
- Department of Nephrology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Yinghong Zhang
- Department of Nephrology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Xiaoyin Zheng
- Department of Clinical Laboratory, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
24
|
Im KI, Kim N, Lee J, Oh UH, Lee HW, Lee DG, Min GJ, Lee R, Lee J, Kim S, Cho SG. SARS-CoV-2-Specific T-Cell as a Potent Therapeutic Strategy against Immune Evasion of Emerging COVID-19 Variants. Int J Mol Sci 2024; 25:10512. [PMID: 39408840 PMCID: PMC11477143 DOI: 10.3390/ijms251910512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Despite advances in vaccination and therapies for coronavirus disease, challenges remain due to reduced antibody longevity and the emergence of virulent variants like Omicron (BA.1) and its subvariants (BA.1.1, BA.2, BA.3, and BA.5). This study explored the potential of adoptive immunotherapy and harnessing the protective abilities using virus-specific T cells (VSTs). Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) VSTs were generated by stimulating donor-derived peripheral blood mononuclear cells with spike, nucleocapsid, and membrane protein peptide mixtures. Phenotypic characterization, including T-cell receptor (TCR) vβ and pentamer analyses, was performed on the ex vivo-expanded cells. We infected human leukocyte antigen (HLA)-partially matched human Calu-3 cells with various authentic SARS-CoV-2 strains in a Biosafety Level 3 facility and co-cultured them with VSTs. VSTs exhibited a diverse TCR vβ repertoire, confirming their ability to target a broad range of SARS-CoV-2 antigens from both the ancestral and mutant strains, including Omicron BA.1 and BA.5. These ex vivo-expanded cells exhibited robust cytotoxicity and low alloreactivity against HLA-partially matched SARS-CoV-2-infected cells. Their cytotoxic effects were consistent across variants, targeting conserved spike and nucleocapsid epitopes. Our findings suggest that third-party partial HLA-matching VSTs could counter immune-escape mechanisms posed by emerging variants of concern.
Collapse
Affiliation(s)
- Keon-Il Im
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Nayoun Kim
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Junseok Lee
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
| | - Ui-Hyeon Oh
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Hye-Won Lee
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Dong-Gun Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.-G.L.); (R.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Gi-June Min
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Department of Hematology, Seoul St. Mary’s Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Raeseok Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.-G.L.); (R.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jinah Lee
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea; (J.L.); (S.K.)
| | - Seungtaek Kim
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea; (J.L.); (S.K.)
| | - Seok-Goo Cho
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
- Department of Hematology, Seoul St. Mary’s Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
25
|
Stepanova E, Isakova-Sivak I, Matyushenko V, Mezhenskaya D, Kudryavtsev I, Kostromitina A, Chistiakova A, Rak A, Bazhenova E, Prokopenko P, Kotomina T, Donina S, Novitskaya V, Sivak K, Karal-Ogly D, Rudenko L. Safety and Immunogenicity Study of a Bivalent Vaccine for Combined Prophylaxis of COVID-19 and Influenza in Non-Human Primates. Vaccines (Basel) 2024; 12:1099. [PMID: 39460266 PMCID: PMC11511058 DOI: 10.3390/vaccines12101099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Influenza and SARS-CoV-2 viruses are two highly variable pathogens. We have developed a candidate bivalent live vaccine based on the strain of licensed A/Leningrad/17-based cold-adapted live attenuated influenza vaccine (LAIV) of H3N2 subtype, which expressed SARS-CoV-2 immunogenic T-cell epitopes. A cassette encoding fragments of S and N proteins of SARS-CoV-2 was inserted into the influenza NA gene using the P2A autocleavage site. In this study, we present the results of preclinical evaluation of the developed bivalent vaccine in a non-human primate model. METHODS Rhesus macaques (Macaca mulatta) (n = 3 per group) were immunized intranasally with 7.5 lg EID50 of the LAIV/CoV-2 bivalent vaccine, a control non-modified H3N2 LAIV or a placebo (chorioallantoic fluid) using a sprayer device, twice, with a 28-day interval. The blood samples were collected at days 0, 3, 28 and 35 for hematological and biochemical assessment. Safety was also assessed by monitoring body weight, body temperature and clinical signs of the disease. Immune responses to influenza virus were assessed both by determining serum antibody titers in hemagglutination inhibition assay, microneutralization assay and IgG ELISA. T-cell responses were measured both to influenza and SARS-CoV-2 antigens using ELISPOT and flow cytometry. Three weeks after the second immunization, animals were challenged with 105 PFU of Delta SARS-CoV-2. The body temperature, weight and challenge virus shedding were monitored for 5 days post-challenge. In addition, virus titers in various organs and histopathology were evaluated on day 6 after SARS-CoV-2 infection. RESULTS There was no toxic effect of the immunizations on the hematological and coagulation hemostasis of animals. No difference in the dynamics of the average weight and thermometry results were found between the groups of animals. Both LAIV and LAIV/CoV-2 variants poorly replicated in the upper respiratory tract of rhesus macaques. Nevertheless, despite this low level of virus shedding, influenza-specific serum IgG responses were detected in the group of monkeys immunized with the LAIV/CoV-2 bivalent but not in the LAIV group. Furthermore, T-cell responses to both influenza and SARS-CoV-2 viruses were detected in the LAIV/CoV-2 vaccine group only. The animals were generally resistant to SARS-CoV-2 challenge, with minimal virus shedding in the placebo and LAIV groups. Histopathological changes in vaccinated animals were decreased compared to the PBS group, suggesting a protective effect of the chimeric vaccine candidate. CONCLUSIONS The candidate bivalent vaccine was safe and immunogenic for non-human primates and warrants its further evaluation in clinical trials.
Collapse
Affiliation(s)
- Ekaterina Stepanova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Irina Isakova-Sivak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Victoria Matyushenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Daria Mezhenskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Igor Kudryavtsev
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Arina Kostromitina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Anna Chistiakova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Alexandra Rak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Ekaterina Bazhenova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Polina Prokopenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Tatiana Kotomina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Svetlana Donina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Vlada Novitskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, Saint-Petersburg 197376, Russia;
| | - Dzhina Karal-Ogly
- Center of Preclinical Research, Research Institute of Medical Primatology, Sochi 354376, Russia;
| | - Larisa Rudenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| |
Collapse
|
26
|
Avelino-Silva VI, Bruhn R, Zurita KG, Deng X, Yu EA, Grebe E, Stone M, Lanteri MC, Spencer BR, Busch MP, Custer B. SARS-CoV-2 antibody levels and long COVID occurrence in blood donors. Transfusion 2024; 64:1719-1731. [PMID: 38984497 DOI: 10.1111/trf.17952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND Long COVID is a common condition lacking consensus definition; determinants remain incompletely understood. Characterizing immune profiles associated with long COVID could support the development of preventive and therapeutic strategies. METHODS We used a survey to investigate blood donors' infection/vaccination history and acute/persistent symptoms following COVID-19. The prevalence of long COVID was evaluated using self-report and an adapted definition from the RECOVER study. We evaluated factors associated with long COVID, focusing on anti-spike and anti-nucleocapsid SARS-CoV-2 antibodies. Lastly, we investigated long COVID clinical subphenotypes using hierarchical clustering. RESULTS Of 33,610 participants, 16,003 (48%) reported having had COVID-19; 1853 (12%) had self-reported long COVID, 685 (4%) met an adapted RECOVER definition, and 2050 (13%) met at least one definition. Higher anti-nucleocapsid levels measured 12-24 weeks post-infection were associated with higher risk of self-reported and RECOVER long COVID. Higher anti-spike IgG levels measured 12-24 weeks post-infection were associated with lower risk of self-reported long COVID. Higher total anti-spike measured 24-48 weeks post-infection was associated with lower risk of RECOVER long COVID. Cluster analysis identified four clinical subphenotypes; patterns included neurological and psychiatric for cluster 1; neurological and respiratory for cluster 2; multi-systemic for cluster 3; and neurological for cluster 4. DISCUSSION Long COVID prevalence in blood donors varies depending on the adopted definition. Anti-SARS-CoV-2 antibodies were time-dependently associated with long COVID; higher anti-nucleocapsid levels were associated with higher risk; and higher anti-spike levels were associated with lower risk of long COVID. Different underlying pathophysiologic mechanisms may be associated with distinct clinical subphenotypes.
Collapse
Affiliation(s)
- Vivian I Avelino-Silva
- Vitalant Research Institute, California, San Francisco, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, California, San Francisco, USA
| | - Roberta Bruhn
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| | - Karla G Zurita
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| | - Xutao Deng
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| | - Elaine A Yu
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| | - Eduard Grebe
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
- South African Centre for Epidemiological Modeling and Analysis (SACEMA), Stellenbosch University, Stellenbosch, South Africa
| | - Mars Stone
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| | - Marion C Lanteri
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
- Creative Testing Solutions, Tempe, Arizona, USA
| | - Bryan R Spencer
- Scientific Affairs, American Red Cross, Rockville, Maryland, USA
| | - Michael P Busch
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| | - Brian Custer
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| |
Collapse
|
27
|
Trinh THK, Tran TD, Pham DL, Nguyen VN, Vu QTT, Pham TD, Nguyen PH, Le MK, Truong DDK, Hoang VA, Huynh N, Ngo DQ, Vuong LN. Characteristics of Immunogenicity against SARS-CoV-2 in a Community-Based Model of Care during the Fourth Wave of COVID-19 Outbreak in Ho Chi Minh City. Yonsei Med J 2024; 65:501-510. [PMID: 39193758 PMCID: PMC11359602 DOI: 10.3349/ymj.2023.0567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/20/2024] [Accepted: 03/25/2024] [Indexed: 08/29/2024] Open
Abstract
PURPOSE Although some immune protection from close contact with individuals who have coronavirus disease 2019 (COVID-19) has been documented, there is limited data on the seroprevalence of antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in individuals who were in lockdown with confirmed COVID-19 cases. This study investigated immunogenicity against SARS-CoV-2 in household members and people who lived near home-quarantined patients with COVID-19. MATERIALS AND METHODS This cross-sectional study was conducted during the community-based care that took place during lockdowns in District 10, Ho Chi Minh City, Vietnam from July to September 2021. SARS-CoV-2 antibody levels were determined in index cases of COVID-19, household contacts, and a no-contact group from the same area. RESULTS A total of 770 participants were included (355 index cases, 103 household contacts, and 312 no contacts). All index cases were unvaccinated, but >90% of individuals in the household and no-contact groups had received ≥1 vaccine dose. SARS-CoV-2 neutralizing antibodies (Nabs) were present in >77% of unvaccinated index cases versus 64%/65.4% in the household/no-contact groups (p=0.001). Antibody concentrations in unvaccinated index cases were significantly higher than those in household contacts and no contacts, with no difference between the latter groups. In all cases, antibody levels declined markedly ≥6 weeks after infection, and failed to persist beyond this time in the household and no-contact groups. CONCLUSION Community-based care may have helped to create community immunogenicity, but Nabs did not persist, highlighting a need for vaccination for all individuals before, or from 6 weeks after, infection with SARS-CoV-2.
Collapse
Affiliation(s)
- Tu Hoang Kim Trinh
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Tuan Diep Tran
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Duy Le Pham
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Vinh Nhu Nguyen
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Quan Tran Thien Vu
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | | | - Phong Hoai Nguyen
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Minh Kieu Le
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | | | - Vu Anh Hoang
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Nghia Huynh
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Dat Quoc Ngo
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Lan Ngoc Vuong
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, Vietnam.
| |
Collapse
|
28
|
Zhou H, Leng P, Wang Y, Yang K, Li C, Ojcius DM, Wang P, Jiang S. Development of T cell antigen-based human coronavirus vaccines against nAb-escaping SARS-CoV-2 variants. Sci Bull (Beijing) 2024; 69:2456-2470. [PMID: 38942698 DOI: 10.1016/j.scib.2024.02.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/15/2023] [Accepted: 02/07/2024] [Indexed: 06/30/2024]
Abstract
Currently approved vaccines have been successful in preventing the severity of COVID-19 and hospitalization. These vaccines primarily induce humoral immune responses; however, highly transmissible and mutated variants, such as the Omicron variant, weaken the neutralization potential of the vaccines, thus, raising serious concerns about their efficacy. Additionally, while neutralizing antibodies (nAbs) tend to wane more rapidly than cell-mediated immunity, long-lasting T cells typically prevent severe viral illness by directly killing infected cells or aiding other immune cells. Importantly, T cells are more cross-reactive than antibodies, thus, highly mutated variants are less likely to escape lasting broadly cross-reactive T cell immunity. Therefore, T cell antigen-based human coronavirus (HCoV) vaccines with the potential to serve as a supplementary weapon to combat emerging SARS-CoV-2 variants with resistance to nAbs are urgently needed. Alternatively, T cell antigens could also be included in B cell antigen-based vaccines to strengthen vaccine efficacy. This review summarizes recent advancements in research and development of vaccines containing T cell antigens or both T and B cell antigens derived from proteins of SARS-CoV-2 variants and/or other HCoVs based on different vaccine platforms.
Collapse
Affiliation(s)
- Hao Zhou
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400016, China.
| | - Ping Leng
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400016, China
| | - Yang Wang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Kaiwen Yang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chen Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - David M Ojcius
- Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, CA 94115, USA
| | - Pengfei Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministry of Education/Ministry of Health/Chinese Academy of Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
29
|
Póvoa da Costa F, de Sarges KML, da Silva R, dos Santos EF, do Nascimento MH, Rodrigues AM, Cantanhede MHD, Rodrigues FBB, Viana MDNDSDA, Leite MDM, de Oliveira CF, Neves PFMD, Pereira Neto GDS, de Brito MTFM, da Silva ALS, Henriques DF, Quaresma JAS, Falcão LFM, Queiroz MAF, Vallinoto IMVC, Vallinoto ACR, Viana GMR, dos Santos EJM. Genetic, Clinical, Epidemiological, and Immunological Profiling of IgG Response Duration after SARS-CoV-2 Infection. Int J Mol Sci 2024; 25:8740. [PMID: 39201427 PMCID: PMC11354850 DOI: 10.3390/ijms25168740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
The IgG response against SARS-CoV-2 infection can persist for over six months (long response; LR). However, among 30% of those infected, the duration can be as short as three months or less (short response; SR). The present study assembled serological data on the anti-SARS-CoV-2 IgG response duration of two previous studies and integrated these results with the plasmatic cytokine levels and genetic profile of 10 immune-relevant SNPs that were also previously published, along with the plasmatic total IgG, IgA, and IgM levels, allowing for the genetic, clinical, immunological, and epidemiological aspects of the post-COVID-19 IgG response duration to be understood. The SR was associated with previous mild acute COVID-19 and with an SNP (rs2228145) in IL6R related to low gene expression. Additionally, among the SR subgroup, no statistically significant Spearman correlations were observed between the plasma levels of IL-17A and the Th17 regulatory cytokines IFN-γ (rs = 0.2399; p = 0.1043), IL-4 (rs = 0.0273; p = 0.8554), and IL-2 (rs = 0.2204; p = 0.1365), while among the LR subgroup, weaker but statistically significant Spearman correlations were observed between the plasma levels of IL-17A and IFN-γ (rs = 0.3873; p = 0.0016), IL-4 (rs = 0.2671; p = 0.0328), and IL-2 (rs = 0.3959; p = 0.0012). These results suggest that the Th17 response mediated by the IL-6 pathway has a role in the prolonged IgG response to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Flávia Póvoa da Costa
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Kevin Matheus Lima de Sarges
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Rosilene da Silva
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Erika Ferreira dos Santos
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Matheus Holanda do Nascimento
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Alice Maciel Rodrigues
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Marcos Henrique Damasceno Cantanhede
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Fabíola Brasil Barbosa Rodrigues
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Maria de Nazaré do Socorro de Almeida Viana
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Mauro de Meira Leite
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Camille Ferreira de Oliveira
- Section of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Health Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 67000-000, Pará, Brazil; (C.F.d.O.); (D.F.H.)
| | - Pablo Fabiano Moura das Neves
- Center of Biological and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil; (P.F.M.d.N.); (J.A.S.Q.); (L.F.M.F.)
| | - Gabriel dos Santos Pereira Neto
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Mioni Thieli Figueiredo Magalhães de Brito
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Andréa Luciana Soares da Silva
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Daniele Freitas Henriques
- Section of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Health Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 67000-000, Pará, Brazil; (C.F.d.O.); (D.F.H.)
| | - Juarez Antônio Simões Quaresma
- Center of Biological and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil; (P.F.M.d.N.); (J.A.S.Q.); (L.F.M.F.)
| | - Luiz Fábio Magno Falcão
- Center of Biological and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil; (P.F.M.d.N.); (J.A.S.Q.); (L.F.M.F.)
| | - Maria Alice Freitas Queiroz
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Izaura Maria Vieira Cayres Vallinoto
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Antonio Carlos Rosário Vallinoto
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Giselle Maria Rachid Viana
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Malaria Basic Research Laboratory, Parasitology Section, Evandro Chagas Institute, Health Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 67000-000, Pará, Brazil
| | - Eduardo José Melo dos Santos
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| |
Collapse
|
30
|
Yan X, Zhao X, Du Y, Wang H, Liu L, Wang Q, Liu J, Wei S. Dynamics of anti-SARS-CoV-2 IgG antibody responses following breakthrough infection and the predicted protective efficacy: A longitudinal community-based population study in China. Int J Infect Dis 2024; 145:107075. [PMID: 38697605 DOI: 10.1016/j.ijid.2024.107075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/05/2024] Open
Abstract
OBJECTIVES To assess the dynamics of the anti-SARS-CoV-2 IgG antibody levels and their efficacy against COVID-19. METHODS We conducted a longitudinal serological analysis of 852 breakthrough COVID-19 infections among the community-based population in Yichang, China. Anti-SARS-CoV-2 IgG levels were measured by chemiluminescence at approximately 3, 4, and 9 months after infection. A linear mixed model predicted IgG antibody decline over 18 months. The effectiveness of antibodies in preventing symptomatic and severe infections was determined using an existing meta-regression model. RESULTS IgG antibodies slowly declined after breakthrough infections. Initially high at around 3 months (339.44 AU/mL, IQR: 262.78-382.95 AU/mL), levels remained significant at 9 months (297.74 AU/mL, IQR: 213.22-360.62 AU/mL). The elderly (≥60 years) had lower antibody levels compared to the young (<20 years) (P < 0.001). The protective efficacy of antibodies against symptomatic and severe infections was lower in the elderly (≥60 years) (78.34% and 86.33%) compared to the young (<20 years) (96.56% and 98.75%) after 1 year. CONCLUSION The study indicated a slow decline in anti-SARS-CoV-2 IgG antibodies, maintaining considerable efficacy for over 1 year. However, lower levels in the elderly suggest reduced protective effects, underscoring the need for age-specific vaccination strategies.
Collapse
Affiliation(s)
- Xiaolong Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zhao
- Center for Disease Control and Prevention, Yichang, Hubei, China
| | - Yin Du
- Center for Disease Control and Prevention, Yichang, Hubei, China
| | - Hao Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianhua Liu
- Center for Disease Control and Prevention, Yichang, Hubei, China
| | - Sheng Wei
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
31
|
Nel I, Ithayakumar A, Blumenthal N, Duneton C, Khourouj VGE, Viala J, Dollfus C, Baudouin V, Guilmin-Crepon S, Theodorou I, Carcelain G. Strategies to determine positive anti-SARS-CoV-2 memory T lymphocyte response during the evolution of an epidemic. J Immunol Methods 2024; 531:113712. [PMID: 38906414 DOI: 10.1016/j.jim.2024.113712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/22/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
During SARS-CoV-2 pandemic, the assessment of immune protection of people at risk of severe infection was an important goal. The appearance of VOCs (Variant of Concern) highlighted the limits of evaluating immune protection through the humoral response. While the humoral response partly loses its neutralizing activity, the anti-SARS-CoV-2 memory T cell response strongly cross protects against VOCs becoming an indispensable tool to assess immune protection. We compared two techniques available in laboratory to evaluate anti-SARS-CoV-2 memory T cell response in a cohort of infected or vaccinated patients with different levels of risk to develop a severe disease: the ELISpot assay and the T-Cell Lymphocyte Proliferation Assay respectively exploring IFNγ production and cell proliferation. We showed that the ELISpot assay detected more anti-Spike memory T cell response than the Lymphocyte Proliferation Assay. We next observed that the use of two different suppliers as antigenic source in the ELISpot assay did not affect the detection of anti-Spike memory T cell response. Finally, we explored a new approach for defining the positivity threshold, using unsupervised mixed Gaussian modeling, challenging the traditional ROC curve used by the supplier. That will be helpful in endemic situation where it could be difficult to recruit "negative" patients.
Collapse
Affiliation(s)
- Isabelle Nel
- Immunology Department, Robert-Debré Hospital, APHP, Paris, France; University Paris Cité, INSERM U976, France
| | | | | | - Charlotte Duneton
- Immunology Department, Robert-Debré Hospital, APHP, Paris, France; University Paris Cité, INSERM U976, France; Pediatric Nephrology Department, Robert-Debré Hospital, APHP, Paris, France
| | | | - Jérôme Viala
- Department of Pediatric Gastroenterology, Robert-Debré Hospital, APHP, Paris, France
| | - Catherine Dollfus
- Pediatric Hematology and Oncology Department, Trousseau Hospital, APHP, Paris, France
| | - Véronique Baudouin
- Pediatric Nephrology Department, Robert-Debré Hospital, APHP, Paris, France
| | - Sophie Guilmin-Crepon
- Clinical Epidemiology Unit, Inserm CIC-EC 1426, Robert-Debré Hospital, APHP, Paris, France
| | | | - Guislaine Carcelain
- Immunology Department, Robert-Debré Hospital, APHP, Paris, France; University Paris Cité, INSERM U976, France.
| |
Collapse
|
32
|
Roohi A, Gharagozlou S. Vitamin D supplementation and calcium: Many-faced gods or nobody in fighting against Corona Virus Disease 2019. Clin Nutr ESPEN 2024; 62:172-184. [PMID: 38901939 DOI: 10.1016/j.clnesp.2024.05.015] [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: 11/15/2023] [Revised: 05/07/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024]
Abstract
In December 2019, Corona Virus Disease 2019 (COVID-19) was first identified and designated as a pandemic in March 2020 due to rapid spread of the virus globally. At the beginning of the pandemic, only a few treatment options, mainly focused on supportive care and repurposing medications, were available. Due to its effects on immune system, vitamin D was a topic of interest during the pandemic, and researchers investigated its potential impact on COVID-19 outcomes. However, the results of studies about the impact of vitamin D on the disease are inconclusive. In the present narrative review, different roles of vitamin D regarding the COVID-19 have been discussed to show that vitamin D supplementation should be recommended carefully.
Collapse
Affiliation(s)
- Azam Roohi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
33
|
Beaudoin-Bussières G, Finzi A. Deciphering Fc-effector functions against SARS-CoV-2. Trends Microbiol 2024; 32:756-768. [PMID: 38365562 DOI: 10.1016/j.tim.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 02/18/2024]
Abstract
Major efforts were deployed to study the antibody response against SARS-CoV-2. Antibodies neutralizing SARS-CoV-2 have been extensively studied in the context of infections, vaccinations, and breakthrough infections. Antibodies, however, are pleiotropic proteins that have many functions in addition to neutralization. These include Fc-effector functions such as antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). Although important to combat viral infections, these Fc-effector functions were less studied in the context of SARS-CoV-2 compared with binding and neutralization. This is partly due to the difficulty in developing reliable assays to measure Fc-effector functions compared to antibody binding and neutralization. Multiple assays have now been developed and can be used to measure different Fc-effector functions. Here, we review these assays and what is known regarding anti-SARS-CoV-2 Fc-effector functions. Overall, this review summarizes and updates our current state of knowledge regarding anti-SARS-CoV-2 Fc-effector functions.
Collapse
Affiliation(s)
- Guillaume Beaudoin-Bussières
- Centre de recherche du CHUM, Montréal, Québec H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Québec H2X 0A9, Canada
| | - Andrés Finzi
- Centre de recherche du CHUM, Montréal, Québec H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Québec H2X 0A9, Canada.
| |
Collapse
|
34
|
Hu Z, López-Muñoz AD, Kosik I, Li T, Callahan V, Brooks K, Yee DS, Holly J, Santos JJS, Castro Brant A, Johnson RF, Takeda K, Zheng ZM, Brenchley JM, Yewdell JW, Fox JM. Recombinant OC43 SARS-CoV-2 spike replacement virus: An improved BSL-2 proxy virus for SARS-CoV-2 neutralization assays. Proc Natl Acad Sci U S A 2024; 121:e2310421121. [PMID: 38976733 PMCID: PMC11260102 DOI: 10.1073/pnas.2310421121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 05/24/2024] [Indexed: 07/10/2024] Open
Abstract
We generated a replication-competent OC43 human seasonal coronavirus (CoV) expressing the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike in place of the native spike (rOC43-CoV2 S). This virus is highly attenuated relative to OC43 and SARS-CoV-2 in cultured cells and animals and is classified as a biosafety level 2 (BSL-2) agent by the NIH biosafety committee. Neutralization of rOC43-CoV2 S and SARS-CoV-2 by S-specific monoclonal antibodies and human sera is highly correlated, unlike recombinant vesicular stomatitis virus-CoV2 S. Single-dose immunization with rOC43-CoV2 S generates high levels of neutralizing antibodies against SARS-CoV-2 and fully protects human ACE2 transgenic mice from SARS-CoV-2 lethal challenge, despite nondetectable replication in respiratory and nonrespiratory organs. rOC43-CoV2 S induces S-specific serum and airway mucosal immunoglobulin A and IgG responses in rhesus macaques. rOC43-CoV2 S has enormous value as a BSL-2 agent to measure S-specific antibodies in the context of a bona fide CoV and is a candidate live attenuated SARS-CoV-2 mucosal vaccine that preferentially replicates in the upper airway.
Collapse
Affiliation(s)
- Zhe Hu
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Alberto Domingo López-Muñoz
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Ivan Kosik
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Tiansheng Li
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Victoria Callahan
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Kelsie Brooks
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Debra S. Yee
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Jaroslav Holly
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Jefferson J. S. Santos
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Ayslan Castro Brant
- Tumor Virus RNA Biology Section, HIV Dynamics and Replication Program, National Cancer Institute, NIH, Frederick, MD21702
| | - Reed F. Johnson
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Kazuyo Takeda
- Microscopy and Imaging Core Facility, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993
| | - Zhi-Ming Zheng
- Tumor Virus RNA Biology Section, HIV Dynamics and Replication Program, National Cancer Institute, NIH, Frederick, MD21702
| | - Jason M. Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Jonathan W. Yewdell
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Julie M. Fox
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| |
Collapse
|
35
|
Lytton SD, Ghosh AK. SARS-CoV-2 Variants and COVID-19 in Bangladesh-Lessons Learned. Viruses 2024; 16:1077. [PMID: 39066238 PMCID: PMC11281597 DOI: 10.3390/v16071077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
The coronavirus infectious disease-2019 (COVID-19) in Bangladesh is a paradigm for how one of the most densely populated countries in the world, with 1270 people per square kilometer, managed to cope with the COVID-19 pandemic under extraordinary circumstances. This review highlights the SARS-CoV-2 variants in Bangladesh and the timeline of their detection in the context of the global experience with the management of vaccination and natural SARS-CoV-2 infection. The motivation to overcome the COVID-19 vaccine dilemma and track Bangladeshi SARS-CoV-2 sub-variants underscores the potential for a low-income country to excel in international medical science, despite having stressed health care services and limited availability of resources for SARS-CoV-2 testing and gene sequencing.
Collapse
Affiliation(s)
| | - Asish Kumar Ghosh
- Department of Virology, Dhaka Medical College Hospital, Dhaka 1000, Bangladesh;
| |
Collapse
|
36
|
Figueroa-Romero A, Atchadé A, Yadouleton A, Fiogbe M, Bonnet E, Yovo E, Accrombessi M, Hounsa S, Paper T, Dupont R, Gaudart J, Le Hesran JY, Massougbodji A, Cottrell G, González R. SARS-CoV-2 seroprevalence among Beninese pregnant women in the third year of the pandemic. BMC Public Health 2024; 24:1762. [PMID: 38956517 PMCID: PMC11221113 DOI: 10.1186/s12889-024-19087-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/10/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Pregnant women are a vulnerable population to COVID-19 given an increased susceptibility to severe SARS-CoV-2 infection and pregnancy complications. However, few SARS-CoV-2 serological surveys have been performed among this population to assess the extent of the infection in sub-Saharan countries. The objectives of this study were to determine SARS-CoV-2 seroprevalence among Beninese pregnant women, to identify spatial seropositivity clusters and to analyse factors associated with the infection. METHODS A cross-sectional study including women in their third trimester of pregnancy attending the antenatal care (ANC) clinics at Allada (south Benin) and Natitingou (north Benin) was conducted. Rapid diagnostic tests (RDT) for detection of IgG/IgM against the SARS-CoV-2 spike protein were performed using capillary blood. Seroprevalence of SARS-CoV-2 antibodies and associations between SARS-CoV-2 serostatus and maternal characteristics were analyzed by multivariate logistic regression. Spatial analyses were performed using the spatial scan statistics to identify spatial clusters of SARS-CoV-2 infection. RESULTS A total of 861 pregnant women were enrolled between May 4 and June 29, 2022. 58/861 (6.7%) participants reported having received COVID-19 vaccine. None of the participants had been diagnosed with COVID-19 during their pregnancy. SARS-CoV-2 antibodies were detected in 607/802 (75.7%; 95% CI 72.56%-78.62%) of unvaccinated participants. Several urban and rural spatial clusters of SARS-CoV-2 cases were identified in Allada and one urban spatial cluster was identified in Natitingou. Unvaccinated participants from Allada with at least one previous morbidity were at a three-times higher risk of presenting SARS-CoV-2 antibodies (OR = 2.89; 95%CI 1.19%-7.00%). CONCLUSION Three out of four pregnant women had SARS-CoV-2 antibodies, suggesting a high virus circulation among pregnant women in Benin, while COVID-19 vaccination coverage was low. Pregnant women with comorbidities may be at increased risk of SARS-CoV-2 infection. This population should be prioritized for COVID-19 diagnosis and vaccination in order to prevent its deleterious effects. TRIAL REGISTRATION NCT06170320 (retrospectively registered on December 21, 2023).
Collapse
Affiliation(s)
- Antía Figueroa-Romero
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic- Universitat de Barcelona, Barcelona, Spain.
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública, CIBERESP, Madrid, Spain.
| | - Aurore Atchadé
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Benin
| | - Anges Yadouleton
- Laboratoire des fièvres hémorragiques virales du Bénin, Ministère de la Santé du Bénin, Cotonou, 01BP918, Bénin
| | - Marc Fiogbe
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Benin
| | - Emmanuel Bonnet
- Institut de recherche pour le développement PRODIG UMR 215, CNRS Université Paris 1 Panthéon- Sorbonne, AgroParisTech 5, cours des Humanités, Aubervilliers, Île-de-France, F-93 322, France
| | - Emmanuel Yovo
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Benin
| | - Manfred Accrombessi
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Benin
- Faculty of Infectious and Tropical Diseases, Disease Control Department, London School of Hygiene & Tropical Medicine, London, UK
- Population Services International, Malaria Department, Country-Based Global Employee, Cotonou, Benin
| | - Sandrine Hounsa
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Benin
| | - Thierry Paper
- Biosynex S.A, 22 boulevard Sebastien Brant, Illkirch Graffenstaden, Strasbourg, F-67400, France
| | - Raphael Dupont
- Biosynex S.A, 22 boulevard Sebastien Brant, Illkirch Graffenstaden, Strasbourg, F-67400, France
| | - Jean Gaudart
- Aix Marseille Univ, IRD, INSERM, SESSTIM, ISSPAM, AP-HM, Hop La Timone, BioSTIC, Biostatistic and ICT, Marseille, France
| | - Jean-Yves Le Hesran
- Institut de Recherche pour le Développement, MERIT UMR216, Université Paris-Cité, Faculté de pharmacie, laboratoire de parasitologie, Paris, France
| | | | - Gilles Cottrell
- Institut de Recherche pour le Développement, MERIT UMR216, Université Paris-Cité, Faculté de pharmacie, laboratoire de parasitologie, Paris, France
| | - Raquel González
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic- Universitat de Barcelona, Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública, CIBERESP, Madrid, Spain
- Department of Medicine, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| |
Collapse
|
37
|
Bauhofer AFL, Ussivane É, Chissaque A, Iahaia F, Pololo R, Campos F, Miranda E, António L, Maholela P, Gatambire A, Djedje M, Ráice F, Gonçalves L, de Deus N, Inlamea O. SARS-CoV-2 in Mozambican primary school-aged children at Maputo City and Province: a cross-sectional study from a low-income country. BMC Pediatr 2024; 24:425. [PMID: 38956534 PMCID: PMC11221092 DOI: 10.1186/s12887-024-04904-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Seroprevalence studies provide information on the true extent of infection and capture demographic and geographic differences, indicating the level of immunity against Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). We sought to provide local evidence of SARS-CoV-2 exposure in school-aged children during in-class teaching in Maputo City and Province, Mozambique. METHODS Between August and November 2022, we performed a cross-sectional study in school-aged children in four schools in rural, peri-urban, and urban areas of Maputo City and Province. A point-of-care test was used to evaluate SARS-CoV-2 antigens and anti-SARS-CoV-2-specific immunoglobulin M (IgM) and immunoglobulin G (IgG) antibodies. Descriptive statistics were used to estimate the prevalence of the antigens and antibodies. Multiple logistic regression models were used to estimate the adjusted odds ratio (AOR) for the factors associated with anti-SARS-CoV-2 antibodies. RESULTS A total of 736 school-aged children were analyzed. The prevalence of the SARS-CoV-2 antigen was 0.5% (4/736). The prevalence of SARS-CoV-2 antigens was 0.0% (0/245), 0.8% (2/240) and 0.8% (2/251), in the rural, peri-urban and urban areas respectively. The overall seroprevalence of the anti-SARS-CoV-2 antibodies (IgG or IgM) was 80.7% (594/736). In rural area anti-SARS-CoV-2 IgG or IgM antibodies were detected in 76.7% (188/245), while in peri-urban area they were detected in 80.0% (192/240) and in urban area they were detected in 85.3% (214/251). In the adjusted logistic regression model, school-aged children from the urban area were more likely to have anti-SARS-CoV-2 IgG or IgM antibodies than were school-aged children from the rural area (adjusted odds ratio: 1.679; 95% CI: 1.060-2.684; p-value = 0.028). CONCLUSIONS During the in-class teaching period, active SARS-CoV-2 cases in school-aged children were observed. More than half of the school-aged children were exposed to SARS-CoV-2, and SARS-CoV-2 was significantly more common in the schools at the urban area than in the school in the rural area at Maputo City and Province.
Collapse
Affiliation(s)
- Adilson Fernando Loforte Bauhofer
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique.
- Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira, Lisboa, Portugal.
| | - Édio Ussivane
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique
| | - Assucênio Chissaque
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique
- Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira, Lisboa, Portugal
| | - Fátima Iahaia
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique
| | - Ramígio Pololo
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique
| | - Fernanda Campos
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique
| | - Emerson Miranda
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique
| | - Luciana António
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique
| | - Plácida Maholela
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique
| | - Aline Gatambire
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique
| | - Marlene Djedje
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique
| | - Fátima Ráice
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique
| | - Luzia Gonçalves
- Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira, Lisboa, Portugal
- Centro de Estatística e Aplicações da Universidade de Lisboa, Lisboa, Portugal
- z-Stat4life, Lisboa, Portugal
| | - Nilsa de Deus
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique
- Departamento de Ciências Biológicas, Universidade Eduardo Mondlane, Maputo, Mozambique
| | - Osvaldo Inlamea
- Instituto Nacional de Saúde (INS), EN1, Bairro da Vila-Parcela, Distrito de Marracuene, Província de Maputo, Mozambique
| |
Collapse
|
38
|
Lee J, Naoe Y, Bang U, Nakagama Y, Saito A, Kido Y, Hotta A. Neutralization sensitivity of SARS-CoV-2 Omicron variants FL.1 and GE.1 by therapeutic antibodies and XBB sera. Virology 2024; 595:110067. [PMID: 38653156 DOI: 10.1016/j.virol.2024.110067] [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: 05/18/2023] [Revised: 02/22/2024] [Accepted: 03/21/2024] [Indexed: 04/25/2024]
Abstract
Two SARS-CoV-2 XBB sub-variants, FL.1 and GE.1, have been increasing in prevalence worldwide, but limited information is available about their ability to evade the immune system. FL.1 and GE.1 are emerging Omicron XBB variants possessing additional mutations in the spike RBD raising concerns of increased neutralization escape. In this study, we assessed the neutralizing ability of eleven FDA-approved monoclonal antibody combinations against different Omicron variants, including BA.2.75, BA.2.76, BA.4/5, XBB.1.5, and CH.1.1. Among the eleven antibodies, Sotrovimab was the only antibody to show broad neutralization ability against XBB.1.5. However, Sotrovimab showed attenuated neutralization efficiency against recently emerging XBB sub-lineages EG.5, FL.1, and GE.1 compared to XBB.1.5. Additionally, XBB.1.5 seropositive convalescent sera displayed lower neutralization activity against EG.5, FL.1, and GE.1. Overall, our findings present enhanced immune evasion capacity of emerging XBB variants and emphasize the importance of continued monitoring of novel variants.
Collapse
Affiliation(s)
- Joseph Lee
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Youichi Naoe
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Uikyu Bang
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Yu Nakagama
- Department of Virology & Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Akatsuki Saito
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan; Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan; Center for Animal Disease Control, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Yasutoshi Kido
- Department of Virology & Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan.
| |
Collapse
|
39
|
Gozlan J, Baron A, Boyd A, Salmona M, Fofana D, Minier M, Gabassi A, Morand-Joubert L, Delaugerre C, Maylin S. Anti-SARS-CoV-2 Neutralizing Responses in Various Populations: Use of a Rapid Surrogate Lateral Flow Assay and Correlations with Anti-RBD Antibody Levels. Life (Basel) 2024; 14:791. [PMID: 39063546 PMCID: PMC11277712 DOI: 10.3390/life14070791] [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: 04/26/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND After the global COVID-19 crisis, understanding post-infectious immunity and vaccine efficacy remains crucial. This study aims to assess anti-SARS-CoV-2 immunity through a quantitative analysis of anti-receptor-binding domain (RBD) antibodies and rapid functional testing of the neutralizing humoral response. METHODS A retrospective analysis was conducted on samples from various cohorts, including partially vaccinated, fully vaccinated, post-COVID/no-vaccination, and post-COVID/vaccination individuals with various immune-competency statuses. The anti-RBD antibodies were measured using an automated chemiluminescence assay, while the neutralizing antibodies' (NAbs') activity was assessed through the lateral flow ichroma COVID-19 nAb test (LFT), a surrogate neutralization assay. RESULTS The analysis revealed various levels of anti-RBD antibodies and seroneutralization responses across cohorts, with the post-COVID/vaccination group demonstrating the most robust protection. A correlation between anti-RBD antibodies and seroneutralization was observed, albeit with varying strength depending on the subgroup analyzed. Longitudinal assessment following natural infection showed an initial surge followed by a decline in both measures. A cutoff of 3.0 log10 BAU/mL was established to predict significant seroneutralization. CONCLUSIONS The ichroma™ COVID-19 nAb test displayed high specificity and emerged as a valuable tool for monitoring anti-SARS-CoV-2 immunity. These findings contribute to understand the antibody response dynamics and underscore the potential of rapid tests in predicting protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Joël Gozlan
- AP-HP, Saint Antoine Hospital, Laboratory of Virology, 75012 Paris, France; (J.G.); (D.F.); (L.M.-J.)
- Reseach’s Department of Saint-Antoine, “Cancer Biology and Therapeutics”, University of Sorbonne, INSERM, 75012 Paris, France
| | - Audrey Baron
- AP-HP, Saint Louis Hospital, Laboratory of Virology, 75010 Paris, France; (A.B.); (M.S.); (M.M.); (A.G.); (C.D.)
| | - Anders Boyd
- Department of Infectious Diseases, Public Health Service of Amsterdam, 1018 WT Amsterdam, The Netherlands;
| | - Maud Salmona
- AP-HP, Saint Louis Hospital, Laboratory of Virology, 75010 Paris, France; (A.B.); (M.S.); (M.M.); (A.G.); (C.D.)
- INSERM U976, University of Paris, 75010 Paris, France
| | - Djeneba Fofana
- AP-HP, Saint Antoine Hospital, Laboratory of Virology, 75012 Paris, France; (J.G.); (D.F.); (L.M.-J.)
| | - Marine Minier
- AP-HP, Saint Louis Hospital, Laboratory of Virology, 75010 Paris, France; (A.B.); (M.S.); (M.M.); (A.G.); (C.D.)
| | - Audrey Gabassi
- AP-HP, Saint Louis Hospital, Laboratory of Virology, 75010 Paris, France; (A.B.); (M.S.); (M.M.); (A.G.); (C.D.)
| | - Laurence Morand-Joubert
- AP-HP, Saint Antoine Hospital, Laboratory of Virology, 75012 Paris, France; (J.G.); (D.F.); (L.M.-J.)
- INSERM, Institut Pierre Louis of Epidémiology and Public Health, University of Sorbonne, 75012 Paris, France
| | - Constance Delaugerre
- AP-HP, Saint Louis Hospital, Laboratory of Virology, 75010 Paris, France; (A.B.); (M.S.); (M.M.); (A.G.); (C.D.)
- INSERM U944, Biology of Emerging Viruses, University of Paris Cité, 75006 Paris, France
| | - Sarah Maylin
- AP-HP, Saint Louis Hospital, Laboratory of Virology, 75010 Paris, France; (A.B.); (M.S.); (M.M.); (A.G.); (C.D.)
| |
Collapse
|
40
|
Claus J, ten Doesschate T, Taks E, Debisarun PA, Smits G, van Binnendijk R, van der Klis F, Verhagen LM, de Jonge MI, Bonten MJM, Netea MG, van de Wijgert JHHM. Determinants of Systemic SARS-CoV-2-Specific Antibody Responses to Infection and to Vaccination: A Secondary Analysis of Randomised Controlled Trial Data. Vaccines (Basel) 2024; 12:691. [PMID: 38932420 PMCID: PMC11209274 DOI: 10.3390/vaccines12060691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
SARS-CoV-2 infections elicit antibodies against the viral spike (S) and nucleocapsid (N) proteins; COVID-19 vaccines against the S-protein only. The BCG-Corona trial, initiated in March 2020 in SARS-CoV-2-naïve Dutch healthcare workers, captured several epidemic peaks and the introduction of COVID-19 vaccines during the one-year follow-up. We assessed determinants of systemic anti-S1 and anti-N immunoglobulin type G (IgG) responses using trial data. Participants were randomised to BCG or placebo vaccination, reported daily symptoms, SARS-CoV-2 test results, and COVID-19 vaccinations, and donated blood for SARS-CoV-2 serology at two time points. In the 970 participants, anti-S1 geometric mean antibody concentrations (GMCs) were much higher than anti-N GMCs. Anti-S1 GMCs significantly increased with increasing number of immune events (SARS-CoV-2 infection or COVID-19 vaccination): 104.7 international units (IU)/mL, 955.0 IU/mL, and 2290.9 IU/mL for one, two, and three immune events, respectively (p < 0.001). In adjusted multivariable linear regression models, anti-S1 and anti-N log10 concentrations were significantly associated with infection severity, and anti-S1 log10 concentration with COVID-19 vaccine type/dose. In univariable models, anti-N log10 concentration was also significantly associated with acute infection duration, and severity and duration of individual symptoms. Antibody concentrations were not associated with long COVID or long-term loss of smell/taste.
Collapse
Affiliation(s)
- Juana Claus
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (J.C.); (T.t.D.); (J.H.H.M.v.d.W.)
| | - Thijs ten Doesschate
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (J.C.); (T.t.D.); (J.H.H.M.v.d.W.)
- Department of Internal Medicine, Jeroen Bosch Ziekenhuis, 5223 GZ Hertogenbosch, The Netherlands
| | - Esther Taks
- Department of Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (E.T.); (P.A.D.); (M.G.N.)
| | - Priya A. Debisarun
- Department of Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (E.T.); (P.A.D.); (M.G.N.)
| | - Gaby Smits
- National Institute of Public Health and the Environment, 3720 BA Bilthoven, The Netherlands; (G.S.); (R.v.B.); (F.v.d.K.)
| | - Rob van Binnendijk
- National Institute of Public Health and the Environment, 3720 BA Bilthoven, The Netherlands; (G.S.); (R.v.B.); (F.v.d.K.)
| | - Fiona van der Klis
- National Institute of Public Health and the Environment, 3720 BA Bilthoven, The Netherlands; (G.S.); (R.v.B.); (F.v.d.K.)
| | - Lilly M. Verhagen
- Department of Paediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Marien I. de Jonge
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Marc J. M. Bonten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (J.C.); (T.t.D.); (J.H.H.M.v.d.W.)
| | - Mihai G. Netea
- Department of Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (E.T.); (P.A.D.); (M.G.N.)
- Department for Genomics & Immunoregulation, Life and Medical Sciences Institute, University of Bonn, 53113 Bonn, Germany
| | - Janneke H. H. M. van de Wijgert
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (J.C.); (T.t.D.); (J.H.H.M.v.d.W.)
| |
Collapse
|
41
|
Oliveira MDS, Carvalho RF, Meurer CM, Rodrigues ÉM, Dias BP, Santos IGCD, Nascimento CA, Rodrigues YM, Santos AJFD, Almeida KDS, de Souza UJB, Campos FS, Ribeiro J, Soares CMDA, Ribeiro Júnior JC. Prevalence, under-reporting, and epidemiological surveillance of COVID-19 in the Araguaína City of Brazil. PLoS One 2024; 19:e0300191. [PMID: 38838042 DOI: 10.1371/journal.pone.0300191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 02/19/2024] [Indexed: 06/07/2024] Open
Abstract
Asymptomatic and underreported individuals remain a source of coronafig disease 2019 (COVID-19) transmission to others. Data on the prevalence and epidemiological factors influencing transmission are fundamental for establishing control measures, especially in vulnerable regions such as the Amazon. This study aimed to determine the point prevalence and active infection of COVID-19 among the population in Araguaína, a Brazilian city located in the Amazon region, analyzed the socioeconomic and behavioral variables of a statistically representative sample of this population using an epidemiological survey, and identify the viral genomic diversity in the region. During the sixth epidemiological week of 2021 (February 8 to 12), samples of 497 inhabitants of the municipality asymptomatic for respiratory syndromes underwent reverse transcription-quantitative polymerase chain reaction and serological tests (immunoglobulin M and immunoglobulin G). A questionnaire collated data on socioeconomic factors, prevention measures, and health status history. The active infection rate was 6.2%, and the prevalence was 13.5% of the study population. Active infection cases were under-reported; each reported positive case represented 14-28 under-reported cases. Lineages P.2, P.1, and B.1.1 were detected. Working from home was a protective factor against the infection, and clinical signs of fever, dry cough, and loss of taste or smell were associated with testing positive (p <0.05). A descriptive analysis of the indicators revealed that the entire population was susceptible to the disease. Intensified vaccination strategies are required regardless of socioeconomic factors, health conditions, and preventive measures. Implementation of objective, comprehensive, and efficient management tools to minimize the spread of COVID-19 in this municipality can serve as a model for other regions of Brazil.
Collapse
Affiliation(s)
- Monike da Silva Oliveira
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, Brazil
- Microbiology Laboratory, Federal University of North Tocantins, Araguaína, Tocantins, Brazil
| | | | - Carolina Merlin Meurer
- Microbiology Laboratory, Federal University of North Tocantins, Araguaína, Tocantins, Brazil
| | - Ézio Machado Rodrigues
- Microbiology Laboratory, Federal University of North Tocantins, Araguaína, Tocantins, Brazil
| | - Bianca Pereira Dias
- Microbiology Laboratory, Federal University of North Tocantins, Araguaína, Tocantins, Brazil
| | | | | | - Yron Moreira Rodrigues
- Microbiology Laboratory, Federal University of North Tocantins, Araguaína, Tocantins, Brazil
| | | | | | | | - Fabrício Souza Campos
- Bioinformatics and Biotechnology Laboratory, Federal University of Tocantins, Gurupi, Tocantins, Brazil
- Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Juliane Ribeiro
- Molecular Biology and Animal Virology Laboratory, State University of Londrina, Londrina, Paraná, Brazil
| | | | | |
Collapse
|
42
|
Kusunoki H, Ohkusa M, Iida R, Saito A, Kawahara M, Ekawa K, Kato N, Motone M, Shimizu H. Increase in antibody titer and change over time associated with severe acute respiratory syndrome coronavirus 2 infection after mRNA vaccination: Consideration of the significance of additional vaccination. Clin Case Rep 2024; 12:e8953. [PMID: 38808194 PMCID: PMC11130231 DOI: 10.1002/ccr3.8953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/19/2024] [Accepted: 05/01/2024] [Indexed: 05/30/2024] Open
Abstract
Key Clinical Message Most Japanese patients naturally infected with COVID-19 were infected after mRNA vaccination, and many maintained high antibody titers due to hybrid immunity. The significance of additional vaccination in hybrid-immunized cases is highly questionable. Abstract Spontaneous infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) after mRNA vaccination causes a marked increase in antibody titer because of the combined effect of vaccine and infection ("hybrid immunity"). In this study, we discuss the significance of the mRNA vaccine booster inoculation that has been repeatedly performed in Japan. We describe the temporal trends of antibody titers in cases in which antibody titers were markedly increased by hybrid immunization. The antibody titer increased with hybrid immunization and tended to decrease with time. However, several cases maintained high antibody titers for approximately 1 year after coronavirus disease 2019 (COVID-19) diagnosis, even without booster vaccination. Most Japanese patients naturally infected with COVID-19 were infected after mRNA vaccination, and many maintained high antibody titers due to hybrid immunity. The significance of additional vaccination in hybrid-immunized cases is highly questionable regarding cost-effectiveness and risk-benefit.
Collapse
Affiliation(s)
- Hiroshi Kusunoki
- Department of Internal MedicineOsaka Dental UniversityHirakataJapan
| | - Michiko Ohkusa
- Department of Laboratory MedicineOsaka Dental University HospitalOsakaJapan
| | - Rie Iida
- Department of Laboratory MedicineOsaka Dental University HospitalOsakaJapan
| | - Ayumi Saito
- Department of Laboratory MedicineOsaka Dental University HospitalOsakaJapan
| | - Mikio Kawahara
- Department of Laboratory MedicineOsaka Dental University HospitalOsakaJapan
| | - Kazumi Ekawa
- Department of Internal MedicineOsaka Dental UniversityHirakataJapan
- Department of Environmental and Preventive MedicineHyogo Medical UniversityNishinomiyaJapan
| | - Nozomi Kato
- Department of Internal MedicineOsaka Dental UniversityHirakataJapan
| | - Masaharu Motone
- Department of Internal MedicineOsaka Dental UniversityHirakataJapan
- Faculty of Health SciencesOsaka Dental UniversityHirakataJapan
| | - Hideo Shimizu
- Department of Internal MedicineOsaka Dental UniversityHirakataJapan
| |
Collapse
|
43
|
Madinga J, Mbala-Kingebeni P, Nkuba-Ndaye A, Baketana-Kinzonzi L, Matungulu-Biyala E, Mutombo-Lupola P, Seghers CA, Smekens T, Ariën KK, Van Damme W, Kalk A, Peeters M, Ahuka-Mundeke S, Muyembe-Tamfum JJ, Vanlerberghe V. COVID-19 seroprevalence cohort survey among health care workers and their household members in Kinshasa, DR Congo, 2020-2022. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2024; 43:74. [PMID: 38824595 PMCID: PMC11144309 DOI: 10.1186/s41043-024-00536-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/17/2024] [Indexed: 06/03/2024]
Abstract
INTRODUCTION Serological surveys offer the most direct measurement to define the immunity status for numerous infectious diseases, such as COVID-19, and can provide valuable insights into understanding transmission patterns. This study describes seroprevalence changes over time in the context of the Democratic Republic of Congo, where COVID-19 case presentation was apparently largely oligo- or asymptomatic, and vaccination coverage remained extremely low. METHODS A cohort of 635 health care workers (HCW) from 5 health zones of Kinshasa and 670 of their household members was interviewed and sampled in 6 rounds between July 2020 and January 2022. At each round, information on risk exposure and a blood sample were collected. Serology was defined as positive when binding antibodies against SARS-CoV-2 spike and nucleocapsid proteins were simultaneously present. RESULTS The SARS-CoV-2 antibody seroprevalence was high at baseline, 17.3% (95% CI 14.4-20.6) and 7.8% (95% CI 5.5-10.8) for HCW and household members, respectively, and fluctuated over time, between 9% and 62.1%. Seropositivity was heterogeneously distributed over the health zones (p < 0.001), ranging from 12.5% (95% CI 6.6-20.8) in N'djili to 33.7% (95% CI 24.6-43.8) in Bandalungwa at baseline for HCW. Seropositivity was associated with increasing rounds adjusted Odds Ratio (aOR) 1.75 (95% CI 1.66-1.85), with increasing age aOR 1.11 (95% CI 1.02-1.20), being a female aOR 1.35 (95% CI 1.10-1.66) and being a HCW aOR 2.38 (95% CI 1.80-3.14). There was no evidence that HCW brought the COVID-19 infection back home, with an aOR of 0.64 (95% CI 0.46-0.91) of seropositivity risk among household members in subsequent surveys. There was seroreversion and seroconversion over time, and HCW had a lower risk of seroreverting than household members (aOR 0.60 (95% CI 0.42-0.86)). CONCLUSION SARS-CoV-2 IgG antibody levels were high and dynamic over time in this African setting with low clinical case rates. The absence of association with health profession or general risk behaviors and with HCW positivity in subsequent rounds in HH members, shows the importance of the time-dependent, and not work-related, force of infection. Cohort seroprevalence estimates in a 'new disease' epidemic seem insufficient to guide policy makers for defining control strategies.
Collapse
Affiliation(s)
- Joule Madinga
- Department of Epidemiology and Global Health, Institut National de Recherche Biomédicale, Faculty of Medicine, University of Kikwit, Kinshasa, Democratic Republic of Congo
| | - Placide Mbala-Kingebeni
- Department of Epidemiology and Global Health, Institut National de Recherche Biomédicale & Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Antoine Nkuba-Ndaye
- Virology Unit, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | - Leonel Baketana-Kinzonzi
- Virology Unit, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | - Elysé Matungulu-Biyala
- Virology Unit, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | - Patrick Mutombo-Lupola
- Department of Epidemiology and Global Health, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | | | - Tom Smekens
- Public Health Department, Institute of Tropical Medicine, Antwerp, Belgium
| | - Kevin K Ariën
- Virology Unit, Institute of Tropical Medicine & Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Wim Van Damme
- Public Health Department, Institute of Tropical Medicine, Antwerp, Belgium
| | - Andreas Kalk
- Deutsche Gesellschaft für Internationale Zusammenarbeit (GIZ), Bonn, Germany
| | - Martine Peeters
- Unit Trans VIHMI, University of Montpellier, IRD/INSERM, Montpellier, France
| | - Steve Ahuka-Mundeke
- Virology Unit, Institut National de Recherche Biomédicale & Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Jean-Jacques Muyembe-Tamfum
- Virology Unit, Institut National de Recherche Biomédicale & Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Veerle Vanlerberghe
- Emerging Infectious Diseases Unit, Public Health Department, Institute of Tropical Medicine, Antwerp, Belgium.
| |
Collapse
|
44
|
Wang P. Potential Usefulness of IgA for the Early Detection of SARS-CoV-2 Infection: Comparison With IgM. Pol J Microbiol 2024; 73:123-130. [PMID: 38905276 PMCID: PMC11192524 DOI: 10.33073/pjm-2024-019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/22/2024] [Indexed: 06/23/2024] Open
Abstract
Serological testing can be a powerful complementary approach to achieve timely diagnosis of severe acute respiratory coronavirus 2 (SARS-CoV-2) infection, along with nucleic acid detection. Immunoglobulin (Ig) A antibodies are less frequently utilized to detect SARS-CoV-2 infection than IgM and IgG antibodies, even though IgA antibodies play an important role in protective immunity against SARS-CoV-2. This review discusses the differences in kinetics and assay performance between IgA and IgM antibodies and the factors influencing antibody responses. It highlights the potential usefulness of analyzing IgA antibodies for the early detection of SARS-CoV-2 infection. The early appearance of IgA and the high sensitivity of IgA-based immunoassays can aid in diagnosing coronavirus disease 2019. However, because of cross-reactivity, it is important to recognize the only moderate specificity of the early detection of SARS-CoV-2 IgA antibodies against spike antigens. Either the analysis of antibodies targeting the nucleocapsid antigen or a combination of antibodies against the nucleocapsid and spike antigens may strengthen the accuracy of serological evaluation.
Collapse
Affiliation(s)
- Pei Wang
- Department of Laboratory Medicine and Blood Transfusion, Jingmen Central Hospital, Jingmen, China
| |
Collapse
|
45
|
Delmonte OM, Oguz C, Dobbs K, Myint-Hpu K, Palterer B, Abers MS, Draper D, Truong M, Kaplan IM, Gittelman RM, Zhang Y, Rosen LB, Snow AL, Dalgard CL, Burbelo PD, Imberti L, Sottini A, Quiros-Roldan E, Castelli F, Rossi C, Brugnoni D, Biondi A, Bettini LR, D'Angio M, Bonfanti P, Anderson MV, Saracino A, Chironna M, Di Stefano M, Fiore JR, Santantonio T, Castagnoli R, Marseglia GL, Magliocco M, Bosticardo M, Pala F, Shaw E, Matthews H, Weber SE, Xirasagar S, Barnett J, Oler AJ, Dimitrova D, Bergerson JRE, McDermott DH, Rao VK, Murphy PM, Holland SM, Lisco A, Su HC, Lionakis MS, Cohen JI, Freeman AF, Snyder TM, Lack J, Notarangelo LD. Perturbations of the T-cell receptor repertoire in response to SARS-CoV-2 in immunocompetent and immunocompromised individuals. J Allergy Clin Immunol 2024; 153:1655-1667. [PMID: 38154666 PMCID: PMC11162338 DOI: 10.1016/j.jaci.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Functional T-cell responses are essential for virus clearance and long-term protection after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, whereas certain clinical factors, such as older age and immunocompromise, are associated with worse outcome. OBJECTIVE We sought to study the breadth and magnitude of T-cell responses in patients with coronavirus disease 2019 (COVID-19) and in individuals with inborn errors of immunity (IEIs) who had received COVID-19 mRNA vaccine. METHODS Using high-throughput sequencing and bioinformatics tools to characterize the T-cell receptor β repertoire signatures in 540 individuals after SARS-CoV-2 infection, 31 IEI recipients of COVID-19 mRNA vaccine, and healthy controls, we quantified HLA class I- and class II-restricted SARS-CoV-2-specific responses and also identified several HLA allele-clonotype motif associations in patients with COVID-19, including a subcohort of anti-type 1 interferon (IFN-1)-positive patients. RESULTS Our analysis revealed that elderly patients with COVID-19 with critical disease manifested lower SARS-CoV-2 T-cell clonotype diversity as well as T-cell responses with reduced magnitude, whereas the SARS-CoV-2-specific clonotypes targeted a broad range of HLA class I- and class II-restricted epitopes across the viral proteome. The presence of anti-IFN-I antibodies was associated with certain HLA alleles. Finally, COVID-19 mRNA immunization induced an increase in the breadth of SARS-CoV-2-specific clonotypes in patients with IEIs, including those who had failed to seroconvert. CONCLUSIONS Elderly individuals have impaired capacity to develop broad and sustained T-cell responses after SARS-CoV-2 infection. Genetic factors may play a role in the production of anti-IFN-1 antibodies. COVID-19 mRNA vaccines are effective in inducing T-cell responses in patients with IEIs.
Collapse
Affiliation(s)
- Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Cihan Oguz
- Integrated Data Sciences Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Katherine Myint-Hpu
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Boaz Palterer
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michael S Abers
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Deborah Draper
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Meng Truong
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | | | | | - Yu Zhang
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Lindsey B Rosen
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Andrew L Snow
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md; Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Md
| | - Clifton L Dalgard
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, Md; The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, Md
| | - Peter D Burbelo
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Md
| | - Luisa Imberti
- Section of Microbiology, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Alessandra Sottini
- Section of Microbiology, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Eugenia Quiros-Roldan
- Department of Infectious and Tropical Diseases, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Francesco Castelli
- Department of Infectious and Tropical Diseases, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Camillo Rossi
- Direzione Sanitaria, ASST Spedali Civili, Brescia, Italy
| | - Duilio Brugnoni
- Laboratorio Analisi Chimico-Cliniche, ASST Spedali Civili, Brescia, Italy
| | - Andrea Biondi
- Pediatric Department and Centro Tettamanti-European Reference Network on Paediatric Cancer, European Reference Network on Haematological Diseases, and European Reference Network on Hereditary Metabolic Disorders, University of Milano-Bicocca-Fondazione MBBM, Monza, Italy
| | - Laura Rachele Bettini
- Pediatric Department and Centro Tettamanti-European Reference Network on Paediatric Cancer, European Reference Network on Haematological Diseases, and European Reference Network on Hereditary Metabolic Disorders, University of Milano-Bicocca-Fondazione MBBM, Monza, Italy
| | - Mariella D'Angio
- Pediatric Department and Centro Tettamanti-European Reference Network on Paediatric Cancer, European Reference Network on Haematological Diseases, and European Reference Network on Hereditary Metabolic Disorders, University of Milano-Bicocca-Fondazione MBBM, Monza, Italy
| | - Paolo Bonfanti
- Department of Infectious Diseases, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Megan V Anderson
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Annalisa Saracino
- Clinic of Infectious Diseases, Azienda Ospedaliero-Universitaria Consorziale Policlinico di Bari, University of Bari, Bari, Italy
| | - Maria Chironna
- Hygiene Section, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Mariantonietta Di Stefano
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, University of Foggia, Foggia, Italy
| | - Jose Ramon Fiore
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, University of Foggia, Foggia, Italy
| | - Teresa Santantonio
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, University of Foggia, Foggia, Italy
| | - Riccardo Castagnoli
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Gian Luigi Marseglia
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Mary Magliocco
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Francesca Pala
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Elana Shaw
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Helen Matthews
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Sarah E Weber
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Sandhya Xirasagar
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Jason Barnett
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Andrew J Oler
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Dimana Dimitrova
- Center for Immuno-Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md
| | - Jenna R E Bergerson
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - David H McDermott
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - V Koneti Rao
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Philip M Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Andrea Lisco
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michail S Lionakis
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Jeffrey I Cohen
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | | | - Justin Lack
- Integrated Data Sciences Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
46
|
Sun B, Chen Z, Feng B, Chen S, Feng S, Wang Q, Niu X, Zhang Z, Zheng P, Lin M, Luo J, Pan Y, Guan S, Zhong N, Chen L. Development of a colloidal gold-based immunochromatographic assay for rapid detection of nasal mucosal secretory IgA against SARS-CoV-2. Front Microbiol 2024; 15:1386891. [PMID: 38881666 PMCID: PMC11177785 DOI: 10.3389/fmicb.2024.1386891] [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: 02/16/2024] [Accepted: 05/13/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction Infection with SARS-CoV-2 begins in the upper respiratory tract and can trigger the production of mucosal spike-specific secretory IgA (sIgA), which provides protection against reinfection. It has been recognized that individuals with high level of nasal spike-specific IgA have a lower risk of reinfection. However, mucosal spike-specific sIgA wanes over time, and different individuals may have various level of spike-specific sIgA and descending kinetics, leading to individual differences in susceptibility to reinfection. A method for detecting spike-specific sIgA in the nasal passage would be valuable for predicting the risk of reinfection so that people at risk can have better preparedness. Methods In this study, we describe the development of a colloidal gold-based immunochromatographic (ICT) strip for detecting SARS-CoV-2 Omicron spike-specific sIgA in nasal mucosal lining fluids (NMLFs). Results The ICT strip was designed to detect 0.125 μg or more spike-specific sIgA in 80 μL of NMLFs collected using a nasal swab. Purified nasal sIgA samples from individuals who recently recovered from an Omicron BA.5 infection were used to demonstrate that this ICT strip can specifically detect spike-specific sIgA. The signal levels positively correlated with neutralizing activities against XBB. Subsequent analysis revealed that people with low or undetectable levels of spike-specific sIgA in the nasal passage were more susceptible to SARS-CoV-2 reinfection. Conclusions This nasal spike-specific sIgA ICT strip provides a non-invasive, rapid, and convenient method to assess the risk of reinfection for achieving precision preparedness.
Collapse
Affiliation(s)
- Baoqing Sun
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
| | - Zhilong Chen
- Xiamen United Institute of Respiratory Health, Xiamen, China
- Xiamen Fortune Bio. Co., Ltd, Xiamen, China
| | - Bo Feng
- Guangzhou Laboratory, Guangzhou, China
| | - Si Chen
- Guangzhou Laboratory, Guangzhou, China
| | | | - Qian Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
| | - Xuefeng Niu
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhengyuan Zhang
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Peiyan Zheng
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ming Lin
- Xiamen United Institute of Respiratory Health, Xiamen, China
- Xiamen Fortune Bio. Co., Ltd, Xiamen, China
| | - Jia Luo
- Xiamen United Institute of Respiratory Health, Xiamen, China
- Xiamen Fortune Bio. Co., Ltd, Xiamen, China
| | - Yingxian Pan
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
47
|
Edner NM, Houghton LP, Ntavli E, Rees-Spear C, Petersone L, Wang C, Fabri A, Elfaki Y, Rueda Gonzalez A, Brown R, Kisand K, Peterson P, McCoy LE, Walker LSK. TIGIT +Tfh show poor B-helper function and negatively correlate with SARS-CoV-2 antibody titre. Front Immunol 2024; 15:1395684. [PMID: 38868776 PMCID: PMC11167088 DOI: 10.3389/fimmu.2024.1395684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/15/2024] [Indexed: 06/14/2024] Open
Abstract
Circulating follicular helper T cells (cTfh) can show phenotypic alterations in disease settings, including in the context of tissue-damaging autoimmune or anti-viral responses. Using severe COVID-19 as a paradigm of immune dysregulation, we have explored how cTfh phenotype relates to the titre and quality of antibody responses. Severe disease was associated with higher titres of neutralising S1 IgG and evidence of increased T cell activation. ICOS, CD38 and HLA-DR expressing cTfh correlated with serum S1 IgG titres and neutralising strength, and interestingly expression of TIGIT by cTfh showed a negative correlation. TIGIT+cTfh expressed increased IFNγ and decreased IL-17 compared to their TIGIT-cTfh counterparts, and showed reduced capacity to help B cells in vitro. Additionally, TIGIT+cTfh expressed lower levels of CD40L than TIGIT-cTfh, providing a potential explanation for their poor B-helper function. These data identify phenotypic changes in polyclonal cTfh that correlate with specific antibody responses and reveal TIGIT as a marker of cTfh with altered function.
Collapse
Affiliation(s)
- Natalie M. Edner
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Luke P. Houghton
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Elisavet Ntavli
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Chloe Rees-Spear
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Lina Petersone
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Chunjing Wang
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Astrid Fabri
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Yassin Elfaki
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Andrea Rueda Gonzalez
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Rachel Brown
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Laura E. McCoy
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Lucy S. K. Walker
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| |
Collapse
|
48
|
Castellano G, Netti GS, Cantaluppi V, Losappio V, Spadaccino F, Ranieri E, Marengo M, Borzumati M, Alfieri C, Stallone G. PMMA dialyzers modulate both humoral and cell-mediate immune response to anti-COVID-19 vaccine (BNT162b2) in a cohort of chronic hemodialyzed patients. Sci Rep 2024; 14:12217. [PMID: 38806543 PMCID: PMC11133365 DOI: 10.1038/s41598-024-62044-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 05/09/2024] [Indexed: 05/30/2024] Open
Abstract
Patients on hemodialysis (HD) have a high risk of death from COVID-19. We evaluated the humoral and cell-mediated immune response to BNT162b2 (Pfizer-BioNTech) vaccine in HD patients, comparing HD with Poly-methyl-methacrylate (PMMA) and HD with Polysulphone (PS). Samples were collected before vaccination (T0) and 14-days after the 2ndvaccine (T2) in a TG (TG, n = 16-Foggia) and in a VG (CG, n = 36-Novara). Anti-SARS-CoV-2-Ig were titrated in the cohort 2-weeks after the 2nddose of vaccine. In the Testing-Group, serum neutralizing antibodies (NAb) were assayed and PBMCs isolated from patients were thawed, counted and stimulated with SARS-CoV-2 IGRA stimulation tube set. All patients had a positive ab-response, except in a case. PMMA-patients had higher levels of anti-SARS-CoV-2 IgG (p = 0.031); VG data confirmed these findings (p < 0.05). NAb evaluation: PMMA patients passed the positive cut-off value, while in PS group only only 1/8 patient did not respond. PMMA patients showed higher percentages of anti-SARS-CoV-2 S1/RBD-Ig after a complete vaccine schedule (p = 0.028). Interferon-gamma release: PMMA patients showed significantly higher release of IFNγ (p = 0.014). The full vaccination course provided sufficient protection against SARS-CoV-2 across the entire cohort, regardless of dialyzer type. After vaccination, PMMA patients show a better immune response, both humoral and cellular, at the end of the vaccination course than PS patients.
Collapse
Affiliation(s)
- Giuseppe Castellano
- Nephrology, Dialysis and Renal Transplantation Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Via Commenda 15, 20122, Milan, Italy.
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| | - Giuseppe Stefano Netti
- Unit of Clinical Pathology, Center for Molecular Medicine and Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Vincenzo Cantaluppi
- Unit of Nephrology and Kidney Transplantation, Department of Translational Medicine, University of Piemonte Orientale (UPO), 17-28100, Novara, Italy
| | - Vincenzo Losappio
- Unit of Nephrology Dialysis and Transplantation, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Federica Spadaccino
- Unit of Clinical Pathology, Center for Molecular Medicine and Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Elena Ranieri
- Unit of Clinical Pathology, Center for Molecular Medicine and Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Marita Marengo
- Unit of Nephrology and Dialysis, Azienda Sanitaria Locale (ASL) CN1, Cuneo, Italy
| | - Maurizio Borzumati
- Unit of Nephrology and Dialysis, ASL VCO, Verbania, Verbano Cusio Ossola, Italy
| | - Carlo Alfieri
- Nephrology, Dialysis and Renal Transplantation Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Via Commenda 15, 20122, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giovanni Stallone
- Unit of Nephrology Dialysis and Transplantation, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
49
|
Mink S, Reimann P, Fraunberger P. Prognostic value of anti-SARS-CoV-2 antibodies: a systematic review. Clin Chem Lab Med 2024; 62:1029-1043. [PMID: 38349073 DOI: 10.1515/cclm-2023-1487] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/02/2024] [Indexed: 04/30/2024]
Abstract
OBJECTIVES Globally, over 772 million cases of COVID-19 have been reported. New variants of interest with corresponding spikes in case numbers continue to be identified. Vulnerable patients, including older adults or patients with severe comorbidities, continue to be at risk. A large body of evidence has been accumulated regarding anti-SARS-CoV-2-antibodies and COVID-19 but the usefulness of antibody measurements remains unclear. This systematic review aims to assess the prognostic value of anti-SARS-CoV-2-antibodies and their usefulness for guiding booster vaccinations. METHODS Studies in English and published between January 2020 and October 2023 were included. Studies that relied on multiparameter-models or comprised fewer than 100 participants were excluded. PubMed and via the WHO COVID-19 research database, Embase and Medline databases were searched. Study selection and quality assessment was conducted independently by two researchers. RESULTS After screening 1,160 studies, 33 studies comprising >30 million individuals were included. Anti-SARS-CoV-2-antibodies were strongly associated with reduced risk of SARS-CoV-2-infection and better outcomes, including mortality. Risk of infection and COVID-19 severity decreased with increasing antibody levels. CONCLUSIONS Anti-SARS-CoV-2-antibodies are useful for early identification of high-risk patients and timely adjustment of therapy. Protective thresholds may be applied to advise booster vaccinations but verification in separate cohorts is required.
Collapse
Affiliation(s)
- Sylvia Mink
- Central Medical Laboratories, Feldkirch, Austria
- Private University in the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
| | - Patrick Reimann
- Private University in the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
- Department of Internal Medicine, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Peter Fraunberger
- Central Medical Laboratories, Feldkirch, Austria
- Private University in the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
| |
Collapse
|
50
|
Taefehshokr N, Lac A, Vrieze AM, Dickson BH, Guo PN, Jung C, Blythe EN, Fink C, Aktar A, Dikeakos JD, Dekaban GA, Heit B. SARS-CoV-2 NSP5 antagonizes MHC II expression by subverting histone deacetylase 2. J Cell Sci 2024; 137:jcs262172. [PMID: 38682259 PMCID: PMC11166459 DOI: 10.1242/jcs.262172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
SARS-CoV-2 interferes with antigen presentation by downregulating major histocompatibility complex (MHC) II on antigen-presenting cells, but the mechanism mediating this process is unelucidated. Herein, analysis of protein and gene expression in human antigen-presenting cells reveals that MHC II is downregulated by the SARS-CoV-2 main protease, NSP5. This suppression of MHC II expression occurs via decreased expression of the MHC II regulatory protein CIITA. CIITA downregulation is independent of the proteolytic activity of NSP5, and rather, NSP5 delivers HDAC2 to the transcription factor IRF3 at an IRF-binding site within the CIITA promoter. Here, HDAC2 deacetylates and inactivates the CIITA promoter. This loss of CIITA expression prevents further expression of MHC II, with this suppression alleviated by ectopic expression of CIITA or knockdown of HDAC2. These results identify a mechanism by which SARS-CoV-2 limits MHC II expression, thereby delaying or weakening the subsequent adaptive immune response.
Collapse
Affiliation(s)
- Nima Taefehshokr
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Alex Lac
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Angela M. Vrieze
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Brandon H. Dickson
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Peter N. Guo
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Catherine Jung
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Eoin N. Blythe
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
- Robarts Research Institute, London, Ontario, CanadaN6A 3K7
| | - Corby Fink
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
- Robarts Research Institute, London, Ontario, CanadaN6A 3K7
| | - Amena Aktar
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
| | - Jimmy D. Dikeakos
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
- Robarts Research Institute, London, Ontario, CanadaN6A 3K7
| | - Gregory A. Dekaban
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
- Robarts Research Institute, London, Ontario, CanadaN6A 3K7
| | - Bryan Heit
- Department of Microbiology and Immunology, and the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, Ontario, CanadaN6A 5C1
- Robarts Research Institute, London, Ontario, CanadaN6A 3K7
| |
Collapse
|