1
|
López MJ, Vazquez MDM, Alvarez-Mon M, Arribas JR, Arana-Arri E, Muñoz P, Navarro-Pérez J, Ramos R, Molto J, Otero-Romero S, Aurrecoechea E, Pomarol R, Bernad L, Esteban I, Pérez-Caballero R, Plana M, Prado JG, Soriano Á. Safety and immunogenicity of PHH-1V booster against SARS-CoV-2 variants, including omicron subvariants: Results from a phase IIb open-label extension study. Hum Vaccin Immunother 2025; 21:2474775. [PMID: 40304691 PMCID: PMC12045571 DOI: 10.1080/21645515.2025.2474775] [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: 11/04/2024] [Revised: 02/20/2025] [Accepted: 02/27/2025] [Indexed: 05/02/2025] Open
Abstract
SARS-CoV-2 vaccination campaigns on current endemic situation would benefit from vaccine alternatives with easy logistics and accessibility, sustained response and cross-reactivity against emerging variants. Herein, safety and immunogenicity of PHH-1V, adjuvanted recombinant RBD-based vaccine, as fourth dose for the most prevalent SARS-CoV-2 variants in Spain in subjects ≥18 years was investigated for 6 months in HIPRA-HH-2 open-label extension study. Subjects received a fourth dose of PHH-1V after either two BNT162b2 doses plus one PHH-1V dose (cohort 1) or three BNT162b2 doses (cohort 2). As regulatory endpoint, neutralization titers were investigated for PHH-1 V as fourth dose vs BNT162b2 as third dose in subjects receiving previous BNT162b2-based regimens. PHH-1 V immunogenicity (GMT) was investigated against Beta, Delta, and Omicron BA.1, BA.4/5 and XBB.1.5 on Days 14, 98 and 182 post-immunization. Two hundred and eighty-eight subjects received PHH-1V. Neutralizing antibodies against Omicron BA.1 at Day 14 significantly increased after the PHH-1V as fourth booster vs the third BNT162b2 booster (GMT ratio 0.43 (95% CI: 0.28; 0.65; p-value < .0001)). PHH-1V fourth booster induced a significant increase in neutralizing titers vs baseline (GMFR on Day 14 [95% CI]: Beta 6.96 [5.23, 9.25]; Delta 6.27 [4.79, 8.22]; Omicron BA.1 9.21 [5.57, 15.21]; Omicron BA.4/5 11.80 [8.29, 16.80]; Omicron XBB.1.5 5.22 [3.97, 6.87]), remaining significantly higher up to 6 months. The most frequent adverse events were injection site pain and fatigue. As conclusion, PHH-1V booster induced sustained humoral and cellular immune response against Beta, Delta variants and cross reactivity against distant Omicron subvariants and could be an appropriate strategy for implementing heterologous vaccination campaigns.
Collapse
Affiliation(s)
- María Jesús López
- Preventive Medicine Unit, Hospital Regional Universitario de Málaga, Málaga, Spain
| | | | - Melchor Alvarez-Mon
- Internal Medicine Unit, Hospital Universitario Príncipe de Asturias, Madrid, Spain
| | - José Ramón Arribas
- Infectious Diseases Unit, Internal Medicine Department, La Paz University Hospital, IdiPAZ, Madrid, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Eunate Arana-Arri
- Scientific Coordinator, Biocruces Bizkaia Health Research Institute, Osakidetza, Barakaldo, Spain.g. Clinical Microbiology, Infectious Diseases and AIDS Group, Instituto de Investigación Sanitaria Hospital Gregorio Marañon, Madrid, Spain
| | - Patricia Muñoz
- Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Gregorio Marañón, Madrid, Spain
- CIBER Enfermedades Respiratorias, CIBERES Group (CB06/06/0058), Madrid, Spain
| | | | - Rafael Ramos
- Vascular Health Research Group, Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Biomedical Research Institute, Girona (IdIBGi), Catalan Institute of Health, Catalonia, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
| | - José Molto
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Infectious Diseases, Fundació Lluita contra les Infeccions, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Susana Otero-Romero
- Preventive Medicine and Epidemiology Department, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Multiple Sclerosis Centre of Catalonia, Department of Neurology/Neuroimmunology, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elena Aurrecoechea
- AIDS Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Roc Pomarol
- AIDS Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Laia Bernad
- Irsicaixa, Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Ignasi Esteban
- AIDS Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Raúl Pérez-Caballero
- Irsicaixa, Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Montserrat Plana
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- AIDS Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Júlia G. Prado
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Irsicaixa, Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Álex Soriano
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
2
|
Peters MQ, Young AL, Stolarczuk JE, Glad M, Layton E, Logue JK, Minkah NK, Chu HY, Englund JA, Sather DN, Seshadri C, Kachikis A, Harrington WE. Infant CD4 T-cell response to SARS-CoV-2 mRNA vaccination is restricted in cytokine production and modified by vaccine manufacturer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646864. [PMID: 40271053 PMCID: PMC12016048 DOI: 10.1101/2025.04.02.646864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Safe and effective vaccines are a key preventative measure to protect infants from SARS-CoV-2 infection and disease. Although mRNA vaccines induce robust antibody titers in infants, little is known about the quality of CD4 T-cell responses induced by vaccination. CD4 T-cell responses are important in orchestrating coordinated immune responses during infection and may help to limit disease severity. METHODS To characterize the CD4 T-cell response to SARS-CoV-2 mRNA vaccination in infants, we sampled blood from 13 infants before and after primary SARS-CoV-2 mRNA vaccine series; samples from 12 historical vaccinated adults were used for comparisons. PBMC were stimulated with Spike peptide pools and the ability of CD4 T-cells to secrete Th1, Th2, and Th17 cytokines was quantified. A measure of polyfunctionality was generated using the COMPASS algorithm. RESULTS We observed a significant increase in CD4 T-cells producing IL-2 (0.01% vs. 0.08%, p=0.04) and TNF-α (0.007% vs. 0.07%, p=0.007) following vaccination in infants but a more muted induction of IFN-γ production (0.01% vs 0.04%, p=0.08). This contrasted with adults, in whom vaccination induced robust production of IFN-γ, IL-2, and TNF-α. Th2 and Th17 responses were limited in both infants and adults. In infants, CD4 T-cell responses post-vaccination were greater in those who received mRNA-1273 versus BNT162b. In contrast to CD4 T-cell responses, Spike-specific IgG titers were similar in infants and adults. CONCLUSIONS These data suggest that infants have restricted induction of cytokine producing CD4 T-cells following SARS-CoV-2 mRNA vaccination relative to adults.
Collapse
|
3
|
Blanco J, Trinité B, Puig‐Barberà J. Rethinking Optimal Immunogens to Face SARS-CoV-2 Evolution Through Vaccination. Influenza Other Respir Viruses 2025; 19:e70076. [PMID: 39871737 PMCID: PMC11773156 DOI: 10.1111/irv.70076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/23/2024] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
SARS-CoV-2, which originated in China in late 2019, quickly fueled the global COVID-19 pandemic, profoundly impacting health and the economy worldwide. A series of vaccines, mostly based on the full SARS-CoV-2 Spike protein, were rapidly developed, showing excellent humoral and cellular responses and high efficacy against both symptomatic infection and severe disease. However, viral evolution and the waning humoral neutralizing responses strongly challenged vaccine long term effectiveness, mainly against symptomatic infection, making necessary a strategy of repeated and updated booster shots. In this repeated vaccination context, antibody repertoire diversification was evidenced, although immune imprinting after booster doses or reinfection was also demonstrated and identified as a major determinant of immunological responses to repeated antigen exposures. Considering that a small domain of the SARS-CoV-2 Spike protein, the receptor binding domain (RBD), is the major target of neutralizing antibodies and concentrates most viral mutations, the following text aims to provide insights into the ongoing debate over the best strategies for vaccine boosters. We address the relevance of developing new booster vaccines that target the evolving RBD, thus focusing on the relevant antigenic sites of the SARS-CoV-2 new variants. A combination of this strategy with immunofusing and computerized approaches could minimize immune imprinting, therefore optimizing neutralizing immune responses and booster vaccine efficacy.
Collapse
Affiliation(s)
- Julià Blanco
- IrsiCaixaBadalonaCataloniaSpain
- Germans Trias i Pujol Research Institute (IGTP)BadalonaCataloniaSpain
- CIBER de Enfermedades InfecciosasMadridSpain
- Chair in Infectious Diseases and Immunity, Faculty of MedicineUniversity of Vic‐Central University of Catalonia (UVic‐UCC)VicCataloniaSpain
| | | | - Joan Puig‐Barberà
- Área de Investigación en VacunasFundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat ValencianaValenciaSpain
| |
Collapse
|
4
|
Smith CP, Hartman RM, Kugler AM, Little V, Baker OR, Fairlie TA, Fernandez RE, Hagen MB, Honie E, Laeyendecker O, Midgley CM, Parker D, Sandoval M, Takahashi S, Hammitt LL, Sutcliffe CG. The Trajectory of Antibody Responses One Year Following SARS-CoV-2 Infection among Indigenous Individuals in the Southwest United States. Viruses 2024; 16:1573. [PMID: 39459907 PMCID: PMC11512241 DOI: 10.3390/v16101573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
SARS-CoV-2 antibody kinetics based on immunologic history is not fully understood. We analyzed anti-spike and anti-nucleocapsid antibody responses following acute infection in a cohort of Indigenous persons. The models of peak concentrations and decay rates estimated that one year after infection, participants would serorevert for anti-nucleocapsid antibodies and remain seropositive for anti-spike antibodies. The peak anti-spike concentrations were higher for individuals vaccinated prior to infection, but the decay rates were similar across immunologic status groups. Children had significantly lower peak anti-spike concentrations than adults. This study affirms the importance of continued vaccination to maintain high levels of immunity in the face of waning immunity.
Collapse
Affiliation(s)
- Claire P. Smith
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Rachel M. Hartman
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Alexa M. Kugler
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Verlena Little
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Owen R. Baker
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Tarayn A. Fairlie
- Division of Coronaviruses and Other Respiratory Viruses, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Reinaldo E. Fernandez
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Melissa B. Hagen
- Division of Coronaviruses and Other Respiratory Viruses, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Elvira Honie
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Oliver Laeyendecker
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Claire M. Midgley
- Division of Coronaviruses and Other Respiratory Viruses, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Dennie Parker
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Marqia Sandoval
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Saki Takahashi
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Laura L. Hammitt
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Catherine G. Sutcliffe
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
5
|
Fung CYJ, Scott M, Lerner-Ellis J, Taher J. Applications of SARS-CoV-2 serological testing: impact of test performance, sample matrices, and patient characteristics. Crit Rev Clin Lab Sci 2024; 61:70-88. [PMID: 37800891 DOI: 10.1080/10408363.2023.2254390] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023]
Abstract
Laboratory testing has been a key tool in managing the SARS-CoV-2 global pandemic. While rapid antigen and PCR testing has proven useful for diagnosing acute SARS-CoV-2 infections, additional testing methods are required to understand the long-term impact of SARS-CoV-2 infections on immune response. Serological testing, a well-documented laboratory practice, measures the presence of antibodies in a sample to uncover information about host immunity. Although proposed applications of serological testing for clinical use have previously been limited, current research into SARS-CoV-2 has shown growing utility for serological methods in these settings. To name a few, serological testing has been used to identify patients with past infections and long-term active disease and to monitor vaccine efficacy. Test utility and result interpretation, however, are often complicated by factors that include poor test sensitivity early in infection, lack of immune response in some individuals, overlying infection and vaccination responses, lack of standardization of antibody titers/levels between instruments, unknown titers that confer immune protection, and large between-individual biological variation following infection or vaccination. Thus, the three major components of this review will examine (1) factors that affect serological test utility: test performance, testing matrices, seroprevalence concerns and viral variants, (2) patient factors that affect serological response: timing of sampling, age, sex, body mass index, immunosuppression and vaccination, and (3) informative applications of serological testing: identifying past infection, immune surveillance to guide health practices, and examination of protective immunity. SARS-CoV-2 serological testing should be beneficial for clinical care if it is implemented appropriately. However, as with other laboratory developed tests, use of SARS-CoV-2 serology as a testing modality warrants careful consideration of testing limitations and evaluation of its clinical utility.
Collapse
Affiliation(s)
- Chun Yiu Jordan Fung
- Lunenfeld Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Mackenzie Scott
- Lunenfeld Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jordan Lerner-Ellis
- Lunenfeld Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer Taher
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Curtis NC, Shin S, Hederman AP, Connor RI, Wieland-Alter WF, Ionov S, Boylston J, Rose J, Sakharkar M, Dorman DB, Dessaint JA, Gwilt LL, Crowley AR, Feldman J, Hauser BM, Schmidt AG, Ashare A, Walker LM, Wright PF, Ackerman ME, Lee J. Characterization of SARS-CoV-2 Convalescent Patients' Serological Repertoire Reveals High Prevalence of Iso-RBD Antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.08.556349. [PMID: 37745524 PMCID: PMC10515772 DOI: 10.1101/2023.09.08.556349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
While our understanding of SARS-CoV-2 pathogenesis and antibody responses following infection and vaccination has improved tremendously since the outbreak in 2019, the sequence identities and relative abundances of the individual constituent antibody molecules in circulation remain understudied. Using Ig-Seq, we proteomically profiled the serological repertoire specific to the whole ectodomain of SARS-CoV-2 prefusion-stabilized spike (S) as well as to the receptor binding domain (RBD) over a 6-month period in four subjects following SARS-CoV-2 infection before SARS-CoV-2 vaccines were available. In each individual, we identified between 59 and 167 unique IgG clonotypes in serum. To our surprise, we discovered that ∼50% of serum IgG specific for RBD did not recognize prefusion-stabilized S (referred to as iso-RBD antibodies), suggesting that a significant fraction of serum IgG targets epitopes on RBD inaccessible on the prefusion-stabilized conformation of S. On the other hand, the abundance of iso-RBD antibodies in nine individuals who received mRNA-based COVID-19 vaccines encoding prefusion-stabilized S was significantly lower (∼8%). We expressed a panel of 12 monoclonal antibodies (mAbs) that were abundantly present in serum from two SARS-CoV-2 infected individuals, and their binding specificities to prefusion-stabilized S and RBD were all in agreement with the binding specificities assigned based on the proteomics data, including 1 iso-RBD mAb which bound to RBD but not to prefusion-stabilized S. 2 of 12 mAbs demonstrated neutralizing activity, while other mAbs were non-neutralizing. 11 of 12 mAbs also bound to S (B.1.351), but only 1 maintained binding to S (B.1.1.529). This particular mAb binding to S (B.1.1.529) 1) represented an antibody lineage that comprised 43% of the individual's total S-reactive serum IgG binding titer 6 months post-infection, 2) bound to the S from a related human coronavirus, HKU1, and 3) had a high somatic hypermutation level (10.9%), suggesting that this antibody lineage likely had been elicited previously by pre-pandemic coronavirus and was re-activated following the SARS-CoV-2 infection. All 12 mAbs demonstrated their ability to engage in Fc-mediated effector function activities. Collectively, our study provides a quantitative overview of the serological repertoire following SARS-CoV-2 infection and the significant contribution of iso-RBD antibodies, demonstrating how vaccination strategies involving prefusion-stabilized S may have reduced the elicitation of iso-RBD serum antibodies which are unlikely to contribute to protection.
Collapse
|
7
|
Gonçalves J, Melro M, Alenquer M, Araújo C, Castro-Neves J, Amaral-Silva D, Ferreira F, Ramalho JS, Charepe N, Serrano F, Pontinha C, Amorim MJ, Soares H. Balance between maternal antiviral response and placental transfer of protection in gestational SARS-CoV-2 infection. JCI Insight 2023; 8:e167140. [PMID: 37490342 PMCID: PMC10544212 DOI: 10.1172/jci.insight.167140] [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: 11/11/2022] [Accepted: 07/19/2023] [Indexed: 07/27/2023] Open
Abstract
The intricate interplay between maternal immune response to SARS-CoV-2 and the transfer of protective factors to the fetus remains unclear. By analyzing mother-neonate dyads from second and third trimester SARS-CoV-2 infections, our study shows that neutralizing antibodies (NAbs) are infrequently detected in cord blood. We uncovered that this is due to impaired IgG-NAb placental transfer in symptomatic infection and to the predominance of maternal SARS-CoV-2 NAbs of the IgA and IgM isotypes, which are prevented from crossing the placenta. Crucially, the balance between maternal antiviral response and transplacental transfer of IgG-NAbs appears to hinge on IL-6 and IL-10 produced in response to SARS-CoV-2 infection. In addition, asymptomatic maternal infection was associated with expansion of anti-SARS-CoV-2 IgM and NK cell frequency. Our findings identify a protective role for IgA/IgM-NAbs in gestational SARS-CoV-2 infection and open the possibility that the maternal immune response to SARS-CoV-2 infection might benefit the neonate in 2 ways, first by skewing maternal immune response toward immediate viral clearance, and second by endowing the neonate with protective mechanisms to curtail horizontal viral transmission in the critical postnatal period, via the priming of IgA/IgM-NAbs to be transferred by the breast milk and via NK cell expansion in the neonate.
Collapse
Affiliation(s)
- Juliana Gonçalves
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Magda Melro
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Marta Alenquer
- Cell Biology of Viral Infection Lab, Gulbenkian Institute of Science, Oeiras, Portugal
- Católica Biomedical Research Centre, Católica Medical School, Portuguese Catholic University, Lisbon, Portugal
| | - Catarina Araújo
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Júlia Castro-Neves
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Daniela Amaral-Silva
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Filipe Ferreira
- Cell Biology of Viral Infection Lab, Gulbenkian Institute of Science, Oeiras, Portugal
- Católica Biomedical Research Centre, Católica Medical School, Portuguese Catholic University, Lisbon, Portugal
| | | | - Nádia Charepe
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- CHRC, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Fátima Serrano
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- CHRC, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| | - Carlos Pontinha
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Maria João Amorim
- Cell Biology of Viral Infection Lab, Gulbenkian Institute of Science, Oeiras, Portugal
- Católica Biomedical Research Centre, Católica Medical School, Portuguese Catholic University, Lisbon, Portugal
| | - Helena Soares
- Human Immunobiology and Pathogenesis Laboratory, iNOVA4Health, Nova Medical School, Faculty of Medical Sciences, Nova University, Lisbon, Portugal
| |
Collapse
|
8
|
Nowill AE, Caruso M, de Campos-Lima PO. T-cell immunity to SARS-CoV-2: what if the known best is not the optimal course for the long run? Adapting to evolving targets. Front Immunol 2023; 14:1133225. [PMID: 37388738 PMCID: PMC10303130 DOI: 10.3389/fimmu.2023.1133225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/11/2023] [Indexed: 07/01/2023] Open
Abstract
Humanity did surprisingly well so far, considering how unprepared it was to respond to the coronavirus disease 2019 (COVID-19) threat. By blending old and ingenious new technology in the context of the accumulated knowledge on other human coronaviruses, several vaccine candidates were produced and tested in clinical trials in record time. Today, five vaccines account for the bulk of the more than 13 billion doses administered worldwide. The ability to elicit biding and neutralizing antibodies most often against the spike protein is a major component of the protection conferred by immunization but alone it is not enough to limit virus transmission. Thus, the surge in numbers of infected individuals by newer variants of concern (VOCs) was not accompanied by a proportional increase in severe disease and death rate. This is likely due to antiviral T-cell responses, whose evasion is more difficult to achieve. The present review helps navigating the very large literature on T cell immunity induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and vaccination. We examine the successes and shortcomings of the vaccinal protection in the light of the emergence of VOCs with breakthrough potential. SARS-CoV-2 and human beings will likely coexist for a long while: it will be necessary to update existing vaccines to improve T-cell responses and attain better protection against COVID-19.
Collapse
Affiliation(s)
- Alexandre E. Nowill
- Integrated Center for Pediatric OncoHaematological Research, State University of Campinas, Campinas, SP, Brazil
| | - Manuel Caruso
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, Québec, QC, Canada
| | - Pedro O. de Campos-Lima
- Boldrini Children’s Center, Campinas, SP, Brazil
- Molecular and Morphofunctional Biology Graduate Program, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
9
|
Zedan HT, Smatti MK, Thomas S, Nasrallah GK, Afifi NM, Hssain AA, Abu Raddad LJ, Coyle PV, Grivel JC, Almaslamani MA, Althani AA, Yassine HM. Assessment of Broadly Reactive Responses in Patients With MERS-CoV Infection and SARS-CoV-2 Vaccination. JAMA Netw Open 2023; 6:e2319222. [PMID: 37389876 PMCID: PMC10314312 DOI: 10.1001/jamanetworkopen.2023.19222] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/25/2023] [Indexed: 07/01/2023] Open
Abstract
Importance In the ongoing COVID-19 pandemic, there remain unanswered questions regarding the nature and importance of the humoral immune response against other coronaviruses. Although coinfection of the Middle East respiratory syndrome coronavirus (MERS-CoV) with the SARS-CoV-2 has not been documented yet, several patients previously infected with MERS-CoV received the COVID-19 vaccine; data describing how preexisting MERS-CoV immunity may shape the response to SARS-CoV-2 following infection or vaccination are lacking. Objective To characterize the cross-reactive and protective humoral responses in patients exposed to both MERS-CoV infection and SARS-CoV-2 vaccination. Design, Setting, and Participants This cohort study involved a total of 18 sera samples collected from 14 patients with MERS-CoV infection before (n = 12) and after (n = 6) vaccination with 2 doses of COVID-19 mRNA vaccine (BNT162b2 or mRNA-1273). Of those patients, 4 had prevaccination and postvaccination samples. Antibody responses to SARS-CoV-2 and MERS-CoV were assessed as well as cross-reactive responses to other human coronaviruses. Main Outcomes and Measures The main outcomes measured were binding antibody responses, neutralizing antibodies, and antibody-dependent cellular cytotoxicity (ADCC) activity. Binding antibodies targeting SARS-CoV-2 main antigens (spike [S], nucleocapsid, and receptor-binding domain) were detected using automated immunoassays. Cross-reactive antibodies with the S1 protein of SARS-CoV, MERS-CoV, and common human coronaviruses were analyzed using a bead-based assay. Neutralizing antibodies (NAbs) against MERS-CoV and SARS-CoV-2 as well as ADCC activity against SARS-CoV-2 were assessed. Results A total of 18 samples were collected from 14 male patients with MERS-CoV infection (mean [SD] age, 43.8 [14.6] years). Median (IQR) duration between primary COVID-19 vaccination and sample collection was 146 (47-189) days. Prevaccination samples had high levels of anti-MERS S1 immunoglobin M (IgM) and IgG (reactivity index ranging from 0.80 to 54.7 for IgM and from 0.85 to 176.3 for IgG). Cross-reactive antibodies with SARS-CoV and SARS-CoV-2 were also detected in these samples. However, cross-reactivity against other coronaviruses was not detected by the microarray assay. Postvaccination samples showed significantly higher levels of total antibodies, IgG, and IgA targeting SARS-CoV-2 S protein compared with prevaccination samples (eg, mean total antibodies: 8955.0 AU/mL; 95% CI, -5025.0 to 22936.0 arbitrary units/mL; P = .002). In addition, significantly higher anti-SARS S1 IgG levels were detected following vaccination (mean reactivity index, 55.4; 95% CI, -9.1 to 120.0; P = .001), suggesting potential cross-reactivity with these coronaviruses. Also, anti-S NAbs were significantly boosted against SARS-CoV-2 (50.5% neutralization; 95% CI, 17.6% to 83.2% neutralization; P < .001) after vaccination. Furthermore, there was no significant increase in antibody-dependent cellular cytotoxicity against SARS-CoV-2 S protein postvaccination. Conclusions and Relevance This cohort study found a significant boost in cross-reactive NAbs in some patients exposed to MERS-CoV and SARS-CoV-2 antigens. These findings suggest that isolation of broadly reactive antibodies from these patients may help guide the development of a pancoronavirus vaccine by targeting cross-reactive epitopes between distinct strains of human coronaviruses.
Collapse
Affiliation(s)
- Hadeel T. Zedan
- Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, Doha, Qatar
| | - Maria K. Smatti
- Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
| | - Swapna Thomas
- Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Gheyath K. Nasrallah
- Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, Doha, Qatar
| | | | - Ali Ait Hssain
- Medical Intensive Care Unit, Hamad Medical Corporation, Doha, Qatar
| | - Laith J. Abu Raddad
- Infectious Disease Epidemiology Group, Department of Population Health Sciences, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Peter V. Coyle
- Virology laboratory, Hamad Medical Corporation, Doha, Qatar
| | | | | | - Asmaa A. Althani
- Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, Doha, Qatar
| | - Hadi M. Yassine
- Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
10
|
Auer A, Bortolami A, Berguido FJ, Bonfante F, Terregino C, Natale A, Fincato A, Colitti B, Rosati S, Lamien CE, Cattoli G. The Luciferase Immunoprecipitation System (LIPS) Targeting the Spike Protein of SARS-CoV-2 Is More Accurate than Nucleoprotein-Based LIPS and ELISAs for Mink Serology. Transbound Emerg Dis 2023; 2023:1318901. [PMID: 40303713 PMCID: PMC12016994 DOI: 10.1155/2023/1318901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/10/2022] [Indexed: 05/02/2025]
Abstract
Since anthropo-zoonotic outbreaks of SARS-CoV-2 have been reported in mink farms, it is important to monitor the seroprevalence within this population. To investigate the accuracy of nucleo (N) or spike (S) protein-based assays to detect anti-SARS-CoV-2 antibodies in animal serum, we compared four assays, two commercial N-based enzyme-linked immunosorbent assays (ELISA) validated for animal sera and two luciferase immunoprecipitation systems (LIPS-N and LIPS-S), to the reference standard plaque reduction neutralisation test (PRNT). Samples included in this study were derived from a naturally infected mink population. For the first time in this study, serum samples of mink were collected over a 307-day period, at different time points, thus providing an overview of performances of four different rapid serological tests over time. The assays were compared by performing a correlation analysis using R2, Spearman's rank-order correlation coefficient, and Fleiss' and Cohen's kappa for analysis of agreement to PRNT, and an UpSet chart was created to visualize the number of shared positive samples between assays. Cohen's kappa test on categorical data showed an excellent agreement between PRNT and LIPS-S, while agreements between PRNT and N-based methods decreased from fair for LIPS-N to poor agreements for the ELISA kits. In addition, LIPS-S revealed the highest number of true-positive SARS-CoV-2 samples compared to N-based methods. Despite an excellent agreement between LIPS-S and PRNT, a weak correlation was detectable between PRNT titres and relative light units. This study shows that the LIPS-S assay can be used for serological surveillance within a naturally exposed mink population, while N-based serological assays are less accurate providing a higher number of false-negative results, especially at a later stage of infection, thus indicating that N antibodies are less persistent in naturally exposed mink. Our findings provide crucial information for veterinarians and competent authorities involved in surveillance and outbreak investigation in wild and farmed minks.
Collapse
Affiliation(s)
- Agathe Auer
- Animal Production and Health Laboratory, Joint FAO and IAEA Centre for Nuclear Applications in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Friedenstrasse 1, A-2444, Seibersdorf, Austria
- Emergency Prevention System (EMPRES), Animal Health Service Food and Agriculture Organization of the United Nations (FAO-UN), Rome, Italy
| | - Alessio Bortolami
- Istituto Zooprofilattico Sperimentale Delle Venezie (IZSVe), Padua, Italy
| | - Francisco J. Berguido
- Animal Production and Health Laboratory, Joint FAO and IAEA Centre for Nuclear Applications in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Friedenstrasse 1, A-2444, Seibersdorf, Austria
| | - Francesco Bonfante
- Istituto Zooprofilattico Sperimentale Delle Venezie (IZSVe), Padua, Italy
| | - Calogero Terregino
- Istituto Zooprofilattico Sperimentale Delle Venezie (IZSVe), Padua, Italy
| | - Alda Natale
- Istituto Zooprofilattico Sperimentale Delle Venezie (IZSVe), Padua, Italy
| | - Alice Fincato
- Istituto Zooprofilattico Sperimentale Delle Venezie (IZSVe), Padua, Italy
| | - Barbara Colitti
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Sergio Rosati
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Charles E. Lamien
- Animal Production and Health Laboratory, Joint FAO and IAEA Centre for Nuclear Applications in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Friedenstrasse 1, A-2444, Seibersdorf, Austria
| | - Giovanni Cattoli
- Animal Production and Health Laboratory, Joint FAO and IAEA Centre for Nuclear Applications in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Friedenstrasse 1, A-2444, Seibersdorf, Austria
| |
Collapse
|
11
|
Carreño JM, Raskin A, Singh G, Tcheou J, Kawabata H, Gleason C, Srivastava K, Vigdorovich V, Dambrauskas N, Gupta SL, González Domínguez I, Martinez JL, Slamanig S, Sather DN, Raghunandan R, Wirachwong P, Muangnoicharoen S, Pitisuttithum P, Wrammert J, Suthar MS, Sun W, Palese P, García-Sastre A, Simon V, Krammer F. An inactivated NDV-HXP-S COVID-19 vaccine elicits a higher proportion of neutralizing antibodies in humans than mRNA vaccination. Sci Transl Med 2023; 15:eabo2847. [PMID: 36791207 DOI: 10.1126/scitranslmed.abo2847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
NDV-HXP-S is a recombinant Newcastle disease virus-based vaccine against SARS-CoV-2, which expresses an optimized (HexaPro) spike protein on its surface. The vaccine can be produced in embryonated chicken eggs using the same process as that used for the production of the vast majority of influenza virus vaccines. Here, we performed a secondary analysis of the antibody responses after vaccination with inactivated NDV-HXP-S in a phase 1 clinical study in Thailand. The SARS-CoV-2 neutralizing and spike protein binding activity of NDV-HXP-S postvaccination serum samples was compared to that of samples from mRNA BNT162b2 (Pfizer) vaccinees. Neutralizing activity of sera from NDV-HXP-S vaccinees was comparable to that of BNT162b2 vaccinees, whereas spike protein binding activity of the NDV-HXP-S vaccinee samples was lower than that of sera obtained from mRNA vaccinees. This led us to calculate ratios between binding and neutralizing antibody titers. Samples from NDV-HXP-S vaccinees had binding to neutralizing activity ratios that were lower than those of BNT162b2 sera, suggesting that NDV-HXP-S vaccination elicits a high proportion of neutralizing antibodies and low non-neutralizing antibody titers. Further analysis showed that, in contrast to mRNA vaccination, which induces strong antibody titers to the receptor binding domain (RBD), the N-terminal domain, and the S2 domain, NDV-HXP-S vaccination induced an RBD-focused antibody response with little reactivity to S2. This finding may explain the high proportion of neutralizing antibodies. In conclusion, vaccination with inactivated NDV-HXP-S induces a high proportion of neutralizing antibodies and absolute neutralizing antibody titers that are comparable to those elicited by mRNA vaccination.
Collapse
Affiliation(s)
- Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Ariel Raskin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Johnstone Tcheou
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Hisaaki Kawabata
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Charles Gleason
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Komal Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Vladimir Vigdorovich
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Nicholas Dambrauskas
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Sneh Lata Gupta
- Department of Pediatrics, Centers for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Emory University, Atlanta, GA 30329, USA.,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Irene González Domínguez
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Jose Luis Martinez
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Stefan Slamanig
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - D Noah Sather
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA.,Department of Pediatrics, University of Washington, Seattle, WA 98109, USA
| | | | - Ponthip Wirachwong
- Government Pharmaceutical Organization, 75/1 Rama VI Road, Ratchathewi, Bangkok 10400, Thailand
| | - Sant Muangnoicharoen
- Vaccine Trial Centre Faculty of Tropical Medicine, Mahidol, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Punnee Pitisuttithum
- Vaccine Trial Centre Faculty of Tropical Medicine, Mahidol, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Jens Wrammert
- Department of Pediatrics, Centers for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Emory University, Atlanta, GA 30329, USA.,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Mehul S Suthar
- Department of Pediatrics, Centers for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Emory University, Atlanta, GA 30329, USA.,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory University, Atlanta, GA 30329, USA
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA.,Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA.,Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA.,Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA.,Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA.,Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA.,Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| |
Collapse
|
12
|
Al-Tamimi M, Tarifi AA, Qaqish A, Abbas MM, Albalawi H, Abu-Raideh J, Salameh M, Khasawneh AI. Immunoglobulins response of COVID-19 patients, COVID-19 vaccine recipients, and random individuals. PLoS One 2023; 18:e0281689. [PMID: 36787317 PMCID: PMC9928079 DOI: 10.1371/journal.pone.0281689] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/29/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND The development of specific immunoglobulins to COVID-19 after natural infection or vaccination has been proposed. The efficacy and dynamics of this response are not clear yet. AIM This study aims to analyze the immunoglobulins response among COVID-19 patients, COVID-19 vaccine recipients and random individuals. METHODS A total of 665 participants including 233 COVID-19 patients, 288 COVID-19 vaccine recipients, and 144 random individuals were investigated for anti-COVID-19 immunoglobulins (IgA, IgG, IgM). RESULTS Among COVID-19 patients, 22.7% had detectable IgA antibodies with a mean of 27.3±57.1 ng/ml, 29.6% had IgM antibodies with a mean of 188.4±666.0 BAU/ml, while 59.2% had IgG antibodies with a mean of 101.7±139.7 BAU/ml. Pfizer-BioNTech vaccine recipients had positive IgG in 99.3% with a mean of 515.5±1143.5 BAU/ml while 85.7% of Sinopharm vaccine recipients had positive IgG with a mean of 170.0±230.0 BAU/ml. Regarding random individuals, 54.9% had positive IgG with a mean of 164.3±214 BAU/ml. The peak IgM response in COVID-19 patients was detected early at 15-22 days, followed by IgG peak at 16-30 days, and IgA peak at 0-60 days. IgM antibodies disappeared at 61-90 days, while IgG and IgA antibodies decreased slowly after the peak and remained detectable up to 300 days. The frequency of IgG positivity among patients was significantly affected by increased age, admission department (inpatient or outpatient), symptoms, need for oxygen therapy, and increased duration between positive COVID-19 RT PCR test and serum sampling (p˂0.05). Positive correlations were noted between different types of immunoglobulins (IgG, IgM, and IgA) among patients. CONCLUSIONS Natural infection and COIVD-19 vaccines provide IgG-mediated immunity. The class, positivity, mean, efficacy, and duration of immunoglobulins response are affected by the mechanism of immunity and host related variables. Random community individuals had detectable COVID-19 IgG at ~55%, far from reaching herd immunity levels.
Collapse
Affiliation(s)
- Mohammad Al-Tamimi
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Amjed A. Tarifi
- Department of Specialized Surgery, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Arwa Qaqish
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Manal M. Abbas
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
- Pharmacological and Diagnostic Research Lab, Al-Ahliyya Amman University, Amman, Jordan
| | - Hadeel Albalawi
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Jumanah Abu-Raideh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Muna Salameh
- Department of Basic Medical Sciences, Faculty of Medicine, AlBalqa Applied University, Alsalt, Jordan
| | - Ashraf I. Khasawneh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| |
Collapse
|
13
|
Djaïleb A, Lavallée É, Parker MF, Cayer MP, Desautels F, de Grandmont MJ, Stuible M, Gervais C, Durocher Y, Trottier S, Boudreau D, Masson JF, Brouard D, Pelletier JN. Assessment of the longitudinal humoral response in non-hospitalized SARS-CoV-2-positive individuals at decentralized sites: Outcomes and concordance. Front Immunol 2023; 13:1052424. [PMID: 36741379 PMCID: PMC9895839 DOI: 10.3389/fimmu.2022.1052424] [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: 10/19/2022] [Accepted: 12/13/2022] [Indexed: 01/22/2023] Open
Abstract
Introduction Early in the COVID-19 pandemic, reagent availability was not uniform, and infrastructure had to be urgently adapted to undertake COVID-19 surveillance. Methods Before the validation of centralized testing, two enzyme-linked immunosorbent assays (ELISA) were established independently at two decentralized sites using different reagents and instrumentation. We compared the results of these assays to assess the longitudinal humoral response of SARS-CoV-2-positive (i.e., PCR-confirmed), non-hospitalized individuals with mild to moderate symptoms, who had contracted SARSCoV-2 prior to the appearance of variants of concern in Québec, Canada. Results The two assays exhibited a high degree of concordance to identify seropositive individuals, thus validating the robustness of the methods. The results also confirmed that serum immunoglobulins persist ≥ 6 months post-infection among non-hospitalized adults and that the antibodies elicited by infection cross-reacted with the antigens from P.1 (Gamma) and B.1.617.2 (Delta) variants of concern. Discussion Together, these results demonstrate that immune surveillance assays can be rapidly and reliably established when centralized testing is not available or not yet validated, allowing for robust immune surveillance.
Collapse
Affiliation(s)
- Abdelhadi Djaïleb
- Département de Chimie, Université de Montréal, Montréal, QC, Canada
- PROTEO, Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des Protéines, Québec, QC, Canada
- Centre en Chimie Verte et Catalyse, Université de Montréal, Montréal, QC, Canada
| | - Étienne Lavallée
- Département de Chimie, Université de Montréal, Montréal, QC, Canada
- PROTEO, Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des Protéines, Québec, QC, Canada
- Centre en Chimie Verte et Catalyse, Université de Montréal, Montréal, QC, Canada
| | - Megan-Faye Parker
- PROTEO, Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des Protéines, Québec, QC, Canada
- Centre en Chimie Verte et Catalyse, Université de Montréal, Montréal, QC, Canada
- Départment de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC, Canada
| | | | | | | | - Matthew Stuible
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, QC, Canada
| | - Christian Gervais
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, QC, Canada
| | - Yves Durocher
- PROTEO, Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des Protéines, Québec, QC, Canada
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, QC, Canada
| | - Sylvie Trottier
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d’Immunologie, Université Laval, Québec, QC, Canada
| | - Denis Boudreau
- Départment de Chimie, Université Laval, Québec, QC, Canada
- Centre d’Optique, Photonique et Laser, Université Laval, Québec, QC, Canada
| | - Jean-Francois Masson
- Département de Chimie, Université de Montréal, Montréal, QC, Canada
- Centre Québécois sur les Matériaux Fonctionnels, Montréal, QC, Canada
- Centre Interdisciplinaire de Recherche sur le Cerveau et l’Apprentissage, Université de Montréal, Montréal, QC, Canada
| | - Danny Brouard
- Héma‐Québec, Affaires Médicales et Innovation, Québec, QC, Canada
| | - Joelle N. Pelletier
- Département de Chimie, Université de Montréal, Montréal, QC, Canada
- PROTEO, Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des Protéines, Québec, QC, Canada
- Centre en Chimie Verte et Catalyse, Université de Montréal, Montréal, QC, Canada
- Départment de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
14
|
Neutralizing Antibody Responses Among Residents and Staff of Long-Term Care Facilities in the State of New Jersey During the First Wave of the COVID-19 Pandemic. J Community Health 2023; 48:50-58. [PMID: 36197535 PMCID: PMC9532818 DOI: 10.1007/s10900-022-01142-z] [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] [Accepted: 09/10/2022] [Indexed: 10/24/2022]
Abstract
Expanding a previous study of the immune response to SARS-CoV-2 in 10 New Jersey long-term care facilities (LTCFs) during the first wave of the pandemic, this study characterized the neutralizing antibody (NAb) response to infection and vaccination among residents and staff. Sera from the original study were tested using the semi-quantitative enzyme-linked immunosorbent cPass neutralization-antibody detection assay. Almost all residents (97.8%) and staff (98.1%) who were positive for IgG S antibody to the spike protein were positive for NAb. In non-vaccinated subjects with a history of infection (positive polymerase chain reaction (PCR) or antigen test), the distribution of mean intervals from infection to serology date was not significantly different for S antibody positives versus negatives. More than 80% of both were positive at 10 months. Similarly, the mean NAb titer for residents and staff was not associated with interval from PCR/antigen positive to serology date, F = 0.1.01, Pr > F = 0.4269 and F = 0.77, Pr > F = 0.6548 respectively. Titers remained high as the interval reached 10 months. In vaccinees who had no history of infection, the NAb titer was near the test maximum when the serum was drawn seven or more days after the second vaccine dose. In staff the mean NAb titer increased significantly as the vaccine number increased from one to two doses, F = 11.69, Pr > F < 0.0001. NAb titers to SARS-CoV-2 in residents and staff of LTCFs were consistently high 10 months after infection and after two doses of vaccine. Ongoing study is needed to determine whether this antibody provides protection as the virus continues to mutate.
Collapse
|
15
|
Violán C, Torán-Monserrat P, Quirant B, Lamonja-Vicente N, Carrasco-Ribelles LA, Chacón C, Manresa-Dominguez JM, Ramos-Roure F, Dacosta-Aguayo R, Palacios-Fernández C, Roso-Llorach A, Pujol A, Ouchi D, Monteagudo M, Montero-Alia P, Garcia-Sierra R, Arméstar F, Doladé M, Prat N, Bonet JM, Clotet B, Blanco I, Boigues-Pons M, Moreno-Millán N, Prado JG, Cáceres EMM. Kinetics of humoral immune response over 17 months of COVID-19 pandemic in a large cohort of healthcare workers in Spain: the ProHEpiC-19 study. BMC Infect Dis 2022; 22:721. [PMID: 36057544 PMCID: PMC9439943 DOI: 10.1186/s12879-022-07696-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/17/2022] [Indexed: 12/15/2022] Open
Abstract
Background Understanding the immune response to the SARS-CoV-2 virus is critical for efficient monitoring and control strategies. The ProHEpic-19 cohort provides a fine-grained description of the kinetics of antibodies after SARS-CoV-2 infection with an exceptional resolution over 17 months. Methods We established a cohort of 769 healthcare workers including healthy and infected with SARS-CoV-2 in northern Barcelona to determine the kinetics of the IgM against the nucleocapsid (N) and the IgG against the N and spike (S) of SARS-CoV-2 in infected healthcare workers. The study period was from 5 May 2020 to 11 November 2021.We used non-linear mixed models to investigate the kinetics of IgG and IgM measured at nine time points over 17 months from the date of diagnosis. The model included factors of time, gender, and disease severity (asymptomatic, mild-moderate, severe-critical) to assess their effects and their interactions. Findings 474 of the 769 participants (61.6%) became infected with SARS-CoV-2. Significant effects of gender and disease severity were found for the levels of all three antibodies. Median IgM(N) levels were already below the positivity threshold in patients with asymptomatic and mild-moderate disease at day 270 after the diagnosis, while IgG(N and S) levels remained positive at least until days 450 and 270, respectively. Kinetic modelling showed a general rise in both IgM(N) and IgG(N) levels up to day 30, followed by a decay with a rate depending on disease severity. IgG(S) levels remained relatively constant from day 15 over time. Interpretation IgM(N) and IgG(N, S) SARS-CoV-2 antibodies showed a heterogeneous kinetics over the 17 months. Only the IgG(S) showed a stable increase, and the levels and the kinetics of antibodies varied according to disease severity. The kinetics of IgM and IgG observed over a year also varied by clinical spectrum can be very useful for public health policies around vaccination criteria in adult population. Funding Regional Ministry of Health of the Generalitat de Catalunya (Call COVID19-PoC SLT16_04; NCT04885478). Supplementary Information The online version contains supplementary material available at 10.1186/s12879-022-07696-6.
Collapse
Affiliation(s)
- Concepción Violán
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Unitat de Suport a la Recerca Metropolitana Nord, Mare de Déu de Guadalupe 2, Planta 1ª, Mataro, 08303, Barcelona, Spain. .,Direcció d'Atenció Primària Metropolitana Nord Institut Català de Salut, Barcelona, Spain. .,Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, S/N, Badalona, 08916, Barcelona, Spain. .,Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.
| | - Pere Torán-Monserrat
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Unitat de Suport a la Recerca Metropolitana Nord, Mare de Déu de Guadalupe 2, Planta 1ª, Mataro, 08303, Barcelona, Spain.,Direcció d'Atenció Primària Metropolitana Nord Institut Català de Salut, Barcelona, Spain.,Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, S/N, Badalona, 08916, Barcelona, Spain.,Department of Medicine, Faculty of Medicine, Universitat de Girona, 17003, Girona, Spain.,Multidisciplinary Research Group in Health and Society GREMSAS (2017 SGR 917), 08007, Barcelona, Spain
| | - Bibiana Quirant
- Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, S/N, Badalona, 08916, Barcelona, Spain.,Cell Biology, Physiology, Immunology Department, FOCIS Center of Excellence-Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Immunology Division, Laboratori Clinic Metropolitana Nord (LCMN), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Noemi Lamonja-Vicente
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Unitat de Suport a la Recerca Metropolitana Nord, Mare de Déu de Guadalupe 2, Planta 1ª, Mataro, 08303, Barcelona, Spain.,Direcció d'Atenció Primària Metropolitana Nord Institut Català de Salut, Barcelona, Spain
| | - Lucía A Carrasco-Ribelles
- Fundació Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Barcelona, Spain
| | - Carla Chacón
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Unitat de Suport a la Recerca Metropolitana Nord, Mare de Déu de Guadalupe 2, Planta 1ª, Mataro, 08303, Barcelona, Spain.,Direcció d'Atenció Primària Metropolitana Nord Institut Català de Salut, Barcelona, Spain
| | - Josep Maria Manresa-Dominguez
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Unitat de Suport a la Recerca Metropolitana Nord, Mare de Déu de Guadalupe 2, Planta 1ª, Mataro, 08303, Barcelona, Spain.,Direcció d'Atenció Primària Metropolitana Nord Institut Català de Salut, Barcelona, Spain.,Immunology Division, Laboratori Clinic Metropolitana Nord (LCMN), Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Nursing, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Francesc Ramos-Roure
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Unitat de Suport a la Recerca Metropolitana Nord, Mare de Déu de Guadalupe 2, Planta 1ª, Mataro, 08303, Barcelona, Spain.,Department of Medicine, Faculty of Medicine, Universitat Autónoma de Barcelona, 08193, Bellaterra, Spain
| | - Rosalia Dacosta-Aguayo
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Unitat de Suport a la Recerca Metropolitana Nord, Mare de Déu de Guadalupe 2, Planta 1ª, Mataro, 08303, Barcelona, Spain.,Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, S/N, Badalona, 08916, Barcelona, Spain
| | - Cristina Palacios-Fernández
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Unitat de Suport a la Recerca Metropolitana Nord, Mare de Déu de Guadalupe 2, Planta 1ª, Mataro, 08303, Barcelona, Spain.,Direcció d'Atenció Primària Metropolitana Nord Institut Català de Salut, Barcelona, Spain
| | - Albert Roso-Llorach
- Fundació Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Barcelona, Spain.,Departament de Pediatria, d'Obstetrícia i Ginecologia i de Medicina Preventiva, Universitat Autónoma de Barcelona, 08193, Bellaterra, Spain
| | - Aleix Pujol
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Unitat de Suport a la Recerca Metropolitana Nord, Mare de Déu de Guadalupe 2, Planta 1ª, Mataro, 08303, Barcelona, Spain.,Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, S/N, Badalona, 08916, Barcelona, Spain
| | - Dan Ouchi
- Fundació Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Barcelona, Spain.,AIDS Research Institute Irsicaixa, Badalona, Spain
| | - Mónica Monteagudo
- Fundació Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Barcelona, Spain
| | - Pilar Montero-Alia
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Unitat de Suport a la Recerca Metropolitana Nord, Mare de Déu de Guadalupe 2, Planta 1ª, Mataro, 08303, Barcelona, Spain.,Direcció d'Atenció Primària Metropolitana Nord Institut Català de Salut, Barcelona, Spain.,Department of Medicine, Faculty of Medicine, Universitat de Girona, 17003, Girona, Spain.,Centre d'Atenció Primària La Riera (Mataró 1), Institut Català de la Salut, Barcelona, Spain
| | - Rosa Garcia-Sierra
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Unitat de Suport a la Recerca Metropolitana Nord, Mare de Déu de Guadalupe 2, Planta 1ª, Mataro, 08303, Barcelona, Spain.,Department of Medicine, Faculty of Medicine, Universitat de Girona, 17003, Girona, Spain.,Department of Nursing, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Fernando Arméstar
- Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Intensive Care Unit, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Maria Doladé
- Clinical and Biochemical Analysis Division, Laboratori Clinic Metropolitana Nord (LCMN), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Nuria Prat
- Direcció d'Atenció Primària Metropolitana Nord Institut Català de Salut, Barcelona, Spain
| | - Josep Maria Bonet
- Direcció d'Atenció Primària Metropolitana Nord Institut Català de Salut, Barcelona, Spain
| | - Bonaventura Clotet
- Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, S/N, Badalona, 08916, Barcelona, Spain.,AIDS Research Institute Irsicaixa, Badalona, Spain.,Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Spain
| | - Ignacio Blanco
- Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Gerència Territorial Metropolitana Nord, Institut Català de la Salut, Barcelona, Spain
| | - Marc Boigues-Pons
- Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, S/N, Badalona, 08916, Barcelona, Spain.,Cell Biology, Physiology, Immunology Department, FOCIS Center of Excellence-Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Immunology Division, Laboratori Clinic Metropolitana Nord (LCMN), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Nemesio Moreno-Millán
- Direcció d'Atenció Primària Metropolitana Nord Institut Català de Salut, Barcelona, Spain
| | - Julia G Prado
- Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, S/N, Badalona, 08916, Barcelona, Spain.,AIDS Research Institute Irsicaixa, Badalona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINF), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Eva María Martínez Cáceres
- Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, S/N, Badalona, 08916, Barcelona, Spain.,Cell Biology, Physiology, Immunology Department, FOCIS Center of Excellence-Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Immunology Division, Laboratori Clinic Metropolitana Nord (LCMN), Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Medicine, Faculty of Medicine, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | | |
Collapse
|
16
|
McCulloch L, Mouat IC, South K, McColl BW, Allan SM, Smith CJ. Stroke-induced changes to immune function and their relevance to increased risk of severe COVID-19 disease. DISCOVERY IMMUNOLOGY 2022; 1:kyac004. [PMID: 38566903 PMCID: PMC10917238 DOI: 10.1093/discim/kyac004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 07/07/2022] [Accepted: 08/01/2022] [Indexed: 04/04/2024]
Abstract
As the COVID-19 pandemic moves towards endemic disease, it remains of key importance to identify groups of individuals vulnerable to severe infection and understand the biological factors that mediate this risk. Stroke patients are at increased risk of developing severe COVID-19, likely due to stroke-induced alterations to systemic immune function. Furthermore, immune responses associated with severe COVID-19 in patients without a history of stroke parallel many of the immune alterations induced by stroke, possibly resulting in a compounding effect that contributes to worsened disease severity. In this review, we discuss the changes to systemic immune function that likely contribute to augmented COVID-19 severity in patients with a history of stroke and the effects of COVID-19 on the immune system that may exacerbate these effects.
Collapse
Affiliation(s)
- Laura McCulloch
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Isobel C Mouat
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Kieron South
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Barry W McColl
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Stuart M Allan
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Craig J Smith
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Greater Manchester Comprehensive Stroke Centre, Manchester Centre for Clinical Neurosciences, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, Salford, UK
| |
Collapse
|
17
|
Barreira GA, Santos EHD, Pereira MFB, Rodrigues KA, Rocha MC, Kanunfre KA, Marques HHDS, Okay TS. Technical performance of a lateral flow immunoassay for detection of anti-SARS-CoV-2 IgG in the outpatient follow-up of non-severe cases and at different times after vaccination: comparison with enzyme and chemiluminescent immunoassays. Rev Inst Med Trop Sao Paulo 2022; 64:e49. [PMID: 35858039 PMCID: PMC9281580 DOI: 10.1590/s1678-9946202264049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/09/2022] [Indexed: 11/22/2022] Open
Abstract
This study assessed the technical performance of a rapid lateral flow immunochromatographic assay (LFIA) for the detection of anti-SARS-CoV-2 IgG and compared LFIA results with chemiluminescent immunoassay (CLIA) results and an in-house enzyme immunoassay (EIA). To this end, a total of 216 whole blood or serum samples from three groups were analyzed: the first group was composed of 68 true negative cases corresponding to blood bank donors, healthy young volunteers, and eight pediatric patients diagnosed with other coronavirus infections. The serum samples from these participants were obtained and stored in a pre-COVID-19 period, thus they were not expected to have COVID-19. In the second group of true positive cases, we chose to replace natural cases of COVID-19 by 96 participants who were expected to have produced anti-SARS-CoV-2 IgG antibodies 30-60 days after the vaccine booster dose. The serum samples were collected on the same day that LFIA were tested either by EIA or CLIA. The third study group was composed of 52 participants (12 adults and 40 children) who did or did not have anti-SARS-CoV-2 IgG antibodies due to specific clinical scenarios. The 12 adults had been vaccinated more than seven months before LFIA testing, and the 40 children had non-severe COVID-19 diagnosed using RT-PCR during the acute phase of infection. They were referred for outpatient follow-up and during this period the serum samples were collected and tested by CLIA and LFIA. All tests were performed by the same healthcare operator and there was no variation of LFIA results when tests were performed on finger prick whole blood or serum samples, so that results were grouped for analysis. LFIA's sensitivity in detecting anti-SARS-CoV-2 IgG antibodies was 90%, specificity 97.6%, efficiency 93%, PPV 98.3%, NPV 86.6%, and likelihood ratio for a positive or a negative result were 37.5 and 0.01 respectively. There was a good agreement (Kappa index of 0.677) between LFIA results and serological (EIA or CLIA) results. In conclusion, LFIA analyzed in this study showed a good technical performance and agreement with reference serological assays (EIA or CLIA), therefore it can be recommended for use in the outpatient follow-up of non-severe cases of COVID-19 and to assess anti-SARS-CoV-2 IgG antibody production induced by vaccination and the antibodies decrease over time. However, LFIAs should be confirmed by using reference serological assays whenever possible.
Collapse
Affiliation(s)
- Gabriel Acca Barreira
- Universidade de São Paulo, Faculdade de Medicina, Instituto de Medicina Tropical de São Paulo, São Paulo, São Paulo, Brazil.,Faculdade Israelita de Ciências da Saúde Albert Einstein, São Paulo, São Paulo, Brazil
| | - Emilly Henrique Dos Santos
- Universidade de São Paulo, Faculdade de Medicina, Instituto de Medicina Tropical de São Paulo, São Paulo, São Paulo, Brazil.,Universidade de São Paulo, Faculdade de Medicina, Departamento de Pediatria, São Paulo, São Paulo, Brazil
| | | | - Karen Alessandra Rodrigues
- Universidade de São Paulo, Faculdade de Medicina, Instituto de Medicina Tropical de São Paulo, São Paulo, São Paulo, Brazil
| | - Mussya Cisotto Rocha
- Universidade de São Paulo, Faculdade de Medicina, Instituto de Medicina Tropical de São Paulo, São Paulo, São Paulo, Brazil
| | - Kelly Aparecida Kanunfre
- Universidade de São Paulo, Faculdade de Medicina, Instituto de Medicina Tropical de São Paulo, São Paulo, São Paulo, Brazil
| | | | - Thelma Suely Okay
- Universidade de São Paulo, Faculdade de Medicina, Instituto de Medicina Tropical de São Paulo, São Paulo, São Paulo, Brazil.,Universidade de São Paulo, Faculdade de Medicina, Departamento de Pediatria, São Paulo, São Paulo, Brazil
| | | |
Collapse
|
18
|
Iwamoto C, Lesteberg KE, Lamb MM, Calvimontes DM, Guo K, Barrett BS, Mickens KL, Duca LM, Monzon J, Chard AN, Guzman G, Barrios E, Rojop N, Arias K, Gomez M, Paiz C, Bolanos GA, Edwards KM, Zielinski Gutierrez E, Azziz-Baumgartner E, Asturias EJ, Santiago ML, Beckham JD, Olson D. High SARS-CoV-2 Seroprevalence and Rapid Neutralizing Antibody Decline among Agricultural Workers in Rural Guatemala, June 2020-March 2021. Vaccines (Basel) 2022; 10:1160. [PMID: 35891324 PMCID: PMC9323551 DOI: 10.3390/vaccines10071160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/16/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Essential agricultural workers work under occupational conditions that may increase the risk of SARS-CoV-2 exposure and transmission. Data from an agricultural worker cohort in Guatemala, and anti-SARS-CoV-2 nucleocapsid IgG (anti-N IgG) testing were used to estimate past infections and analyze risk factors associated with seropositivity at enrollment and association with SARS-CoV-2 infection. The stability of neutralizing antibody (NAb) responses were assessed in a subset of participants. The adjusted relative risk (aRR) for seroprevalence at enrollment was estimated accounting for correlations within worksites. At enrollment, 616 (46.2%) of 1334 (93.2%) participants had anti-N IgG results indicating prior SARS-CoV-2 infection. A cough ≤ 10 days prior to enrollment (aRR = 1.28, 95% CI: 1.13−1.46) and working as a packer (aRR = 2.00, 95% CI: 1.67−2.38) or packing manager within the plants (aRR = 1.82, 95% CI: 1.36−2.43) were associated with increased risk of seropositivity. COVID-19 incidence density among seronegative workers was 2.3/100 Person-Years (P-Y), higher than seropositive workers (0.4/100 P-Y). Most workers with follow-up NAb testing (65/77, 84%) exhibited a 95% average decrease in NAb titers in <6 months. While participants seropositive at baseline were less likely to experience a symptomatic SARS-CoV-2 infection during follow-up, NAb titers rapidly waned, underscoring the need for multipronged COVID-19 prevention strategies in the workplace, including vaccination.
Collapse
Affiliation(s)
- Chelsea Iwamoto
- Influenza Division, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA; (L.M.D.); (A.N.C.); (E.A.-B.)
| | - Kelsey E. Lesteberg
- Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine, 13001 E. 17th Pl, Aurora, CO 80045, USA; (K.E.L.); (K.G.); (B.S.B.); (K.L.M.); (M.L.S.); (J.D.B.)
| | - Molly M. Lamb
- Department of Epidemiology and Center for Global Health, Colorado School of Public Health, 13199 E. Montview Blvd, Aurora, CO 80045, USA; (M.M.L.); (E.J.A.); (D.O.)
| | - Diva M. Calvimontes
- Center for Human Development, Fundación para la Salud Integral de los Guatemaltecos, FSIG, Km 30 carretera de Coatepeque a Chiquirines Caballo Blanco, Retalhuleu 11010, Guatemala; (D.M.C.); (G.G.); (E.B.); (N.R.); (K.A.); (M.G.); (C.P.); (G.A.B.)
- La Comisión Presidencial de Atención a la Emergencia COVID-19 (Coprecovid), Guatemala City 01010, Guatemala
| | - Kejun Guo
- Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine, 13001 E. 17th Pl, Aurora, CO 80045, USA; (K.E.L.); (K.G.); (B.S.B.); (K.L.M.); (M.L.S.); (J.D.B.)
| | - Bradley S. Barrett
- Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine, 13001 E. 17th Pl, Aurora, CO 80045, USA; (K.E.L.); (K.G.); (B.S.B.); (K.L.M.); (M.L.S.); (J.D.B.)
| | - Kaylee L. Mickens
- Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine, 13001 E. 17th Pl, Aurora, CO 80045, USA; (K.E.L.); (K.G.); (B.S.B.); (K.L.M.); (M.L.S.); (J.D.B.)
| | - Lindsey M. Duca
- Influenza Division, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA; (L.M.D.); (A.N.C.); (E.A.-B.)
| | - Jose Monzon
- Centers for Disease Control and Prevention, Division of Global Health Protection (CDC-DGHP), 1600 Clifton Rd., Atlanta, GA 30329, USA; (J.M.); (E.Z.G.)
| | - Anna N. Chard
- Influenza Division, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA; (L.M.D.); (A.N.C.); (E.A.-B.)
| | - Gerber Guzman
- Center for Human Development, Fundación para la Salud Integral de los Guatemaltecos, FSIG, Km 30 carretera de Coatepeque a Chiquirines Caballo Blanco, Retalhuleu 11010, Guatemala; (D.M.C.); (G.G.); (E.B.); (N.R.); (K.A.); (M.G.); (C.P.); (G.A.B.)
| | - Edgar Barrios
- Center for Human Development, Fundación para la Salud Integral de los Guatemaltecos, FSIG, Km 30 carretera de Coatepeque a Chiquirines Caballo Blanco, Retalhuleu 11010, Guatemala; (D.M.C.); (G.G.); (E.B.); (N.R.); (K.A.); (M.G.); (C.P.); (G.A.B.)
| | - Neudy Rojop
- Center for Human Development, Fundación para la Salud Integral de los Guatemaltecos, FSIG, Km 30 carretera de Coatepeque a Chiquirines Caballo Blanco, Retalhuleu 11010, Guatemala; (D.M.C.); (G.G.); (E.B.); (N.R.); (K.A.); (M.G.); (C.P.); (G.A.B.)
| | - Kareen Arias
- Center for Human Development, Fundación para la Salud Integral de los Guatemaltecos, FSIG, Km 30 carretera de Coatepeque a Chiquirines Caballo Blanco, Retalhuleu 11010, Guatemala; (D.M.C.); (G.G.); (E.B.); (N.R.); (K.A.); (M.G.); (C.P.); (G.A.B.)
| | - Melissa Gomez
- Center for Human Development, Fundación para la Salud Integral de los Guatemaltecos, FSIG, Km 30 carretera de Coatepeque a Chiquirines Caballo Blanco, Retalhuleu 11010, Guatemala; (D.M.C.); (G.G.); (E.B.); (N.R.); (K.A.); (M.G.); (C.P.); (G.A.B.)
| | - Claudia Paiz
- Center for Human Development, Fundación para la Salud Integral de los Guatemaltecos, FSIG, Km 30 carretera de Coatepeque a Chiquirines Caballo Blanco, Retalhuleu 11010, Guatemala; (D.M.C.); (G.G.); (E.B.); (N.R.); (K.A.); (M.G.); (C.P.); (G.A.B.)
| | - Guillermo Antonio Bolanos
- Center for Human Development, Fundación para la Salud Integral de los Guatemaltecos, FSIG, Km 30 carretera de Coatepeque a Chiquirines Caballo Blanco, Retalhuleu 11010, Guatemala; (D.M.C.); (G.G.); (E.B.); (N.R.); (K.A.); (M.G.); (C.P.); (G.A.B.)
| | - Kathryn M. Edwards
- Division of Infectious Disease, Department of Pediatrics, Vanderbilt University School of Medicine, 2200 Children’s Way, 6th Floor, Nashville, TN 37232, USA;
| | - Emily Zielinski Gutierrez
- Centers for Disease Control and Prevention, Division of Global Health Protection (CDC-DGHP), 1600 Clifton Rd., Atlanta, GA 30329, USA; (J.M.); (E.Z.G.)
| | - Eduardo Azziz-Baumgartner
- Influenza Division, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA; (L.M.D.); (A.N.C.); (E.A.-B.)
| | - Edwin J. Asturias
- Department of Epidemiology and Center for Global Health, Colorado School of Public Health, 13199 E. Montview Blvd, Aurora, CO 80045, USA; (M.M.L.); (E.J.A.); (D.O.)
- Center for Human Development, Fundación para la Salud Integral de los Guatemaltecos, FSIG, Km 30 carretera de Coatepeque a Chiquirines Caballo Blanco, Retalhuleu 11010, Guatemala; (D.M.C.); (G.G.); (E.B.); (N.R.); (K.A.); (M.G.); (C.P.); (G.A.B.)
- La Comisión Presidencial de Atención a la Emergencia COVID-19 (Coprecovid), Guatemala City 01010, Guatemala
- Division of Infectious Disease, Department of Pediatrics, University of Colorado School of Medicine, 13123 E. 16th Ave., Aurora, CO 80045, USA
| | - Mario L. Santiago
- Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine, 13001 E. 17th Pl, Aurora, CO 80045, USA; (K.E.L.); (K.G.); (B.S.B.); (K.L.M.); (M.L.S.); (J.D.B.)
| | - J. David Beckham
- Division of Infectious Diseases, Department of Medicine, University of Colorado School of Medicine, 13001 E. 17th Pl, Aurora, CO 80045, USA; (K.E.L.); (K.G.); (B.S.B.); (K.L.M.); (M.L.S.); (J.D.B.)
| | - Daniel Olson
- Department of Epidemiology and Center for Global Health, Colorado School of Public Health, 13199 E. Montview Blvd, Aurora, CO 80045, USA; (M.M.L.); (E.J.A.); (D.O.)
- Center for Human Development, Fundación para la Salud Integral de los Guatemaltecos, FSIG, Km 30 carretera de Coatepeque a Chiquirines Caballo Blanco, Retalhuleu 11010, Guatemala; (D.M.C.); (G.G.); (E.B.); (N.R.); (K.A.); (M.G.); (C.P.); (G.A.B.)
- Division of Infectious Disease, Department of Pediatrics, University of Colorado School of Medicine, 13123 E. 16th Ave., Aurora, CO 80045, USA
| |
Collapse
|
19
|
Jeewandara C, Jayampathi KACS, Ranasinghe T, Aberathna IS, Gunasekara B, Danasekara S, Nimasha T, Kuruppu H, Dissanayake O, Gamalath N, Ekanayake D, Jayamali J, Somathilake G, Guruge D, Wijayamuni R, Kamaladasa A, Ogg GS, Malavige GN. Antibody responses to Sinopharm/BBIBP-CorV in pregnant mothers in Sri Lanka. PLOS GLOBAL PUBLIC HEALTH 2022; 2:e0000607. [PMID: 36962461 PMCID: PMC10022122 DOI: 10.1371/journal.pgph.0000607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/16/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND There are limited data regarding the safety and immunogenicity of the Sinopharm/BBIBP-CorV vaccine in pregnancy. Therefore, we sought to investigate the antibody responses and maternal and fetal adverse events following this vaccine in pregnant mothers in Sri Lanka. METHODS AND FINDINGS SARS-CoV-2 receptor binding domain (RBD) specific total antibodies and ACE2 blocking antibodies were measured by ELISA in pregnant mothers (n = 94) who received the vaccine in the first (n = 2), second (n = 57) and third (n = 33) trimester of pregnancy. Data regarding adverse events and fetal and maternal outcomes were obtained from the women once they delivered. No adverse maternal or fetal complications reported such as miscarriage, thrombotic events, hypertensive disorders, fetal death, preterm delivery, or congenital anomalies were reported. 58/94 (61.7%) had RBD binding antibodies and were found to be seropositive at the time of recruitment. All women seroconverted after the second dose and 31/36 previously uninfected women and 57/58 previously infected women gave a positive response to ACE2 blocking antibodies. The RBD binding antibody levels (p = 0.0002) and ACE2 blocking antibodies (p<0.0001) were significantly higher in previously infected individuals post-second dose compared to uninfected individuals. CONCLUSIONS The Sinopharm/ BBIBP-CorV vaccine appeared safe and induced high seroconversion rates and ACE2 blocking antibodies in pregnant mothers in the second and third trimester in pregnancy. However, the RBD binding antibodies and ACE2 blocking antibodies post-second dose were significantly higher in previously infected pregnant mothers post-second dose, suggesting that two doses of the vaccine are likely to be less immunogenic in previously unexposed individuals.
Collapse
Affiliation(s)
- Chandima Jeewandara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | | | - Thushali Ranasinghe
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Inoka Sepali Aberathna
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Banuri Gunasekara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Saubhagya Danasekara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Thashmi Nimasha
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Heshan Kuruppu
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Osanda Dissanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Nayanathara Gamalath
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Dinithi Ekanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Jewantha Jayamali
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Gayasha Somathilake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | | | | | - Achala Kamaladasa
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Graham S. Ogg
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Gathsaurie Neelika Malavige
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Hastert FD, Henss L, von Rhein C, Gerbeth J, Wieters I, Borgans F, Khodamoradi Y, Zacharowski K, Rohde G, Vehreschild MJ, Schnierle BS. Longitudinal Analysis of Coronavirus-Neutralizing Activity in COVID-19 Patients. Viruses 2022; 14:882. [PMID: 35632624 PMCID: PMC9144377 DOI: 10.3390/v14050882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic has now been continuing for more than two years. The infection causes COVID-19, a disease of the respiratory and cardiovascular system of variable severity. Here, the humoral immune response of 80 COVID-19 patients from the University Hospital Frankfurt/Main, Germany, was characterized longitudinally. The SARS-CoV-2 neutralization activity of serum waned over time. The neutralizing potential of serum directed towards the human alpha-coronavirus NL-63 (NL63) also waned, indicating that no cross-priming against alpha-coronaviruses occurred. A subset of the recovered patients (n = 13) was additionally vaccinated with the mRNA vaccine Comirnaty. Vaccination increased neutralization activity against SARS-CoV-2 wild-type (WT), Delta, and Omicron, although Omicron-specific neutralization was not detectable prior to vaccination. In addition, the vaccination induced neutralizing antibodies against the more distantly related SARS-CoV-1 but not against NL63. The results indicate that although SARS-CoV-2 humoral immune responses induced by infection wane, vaccination induces a broad neutralizing activity against multiple SARS-CoVs, but not to the common cold alpha-coronavirus NL63.
Collapse
Affiliation(s)
- Florian D. Hastert
- Department of Virology, Paul-Ehrlich-Institut, Paul-Ehrlich Strass 51-59, 63225 Langen, Germany; (F.D.H.); (L.H.); (C.v.R.); (J.G.)
| | - Lisa Henss
- Department of Virology, Paul-Ehrlich-Institut, Paul-Ehrlich Strass 51-59, 63225 Langen, Germany; (F.D.H.); (L.H.); (C.v.R.); (J.G.)
| | - Christine von Rhein
- Department of Virology, Paul-Ehrlich-Institut, Paul-Ehrlich Strass 51-59, 63225 Langen, Germany; (F.D.H.); (L.H.); (C.v.R.); (J.G.)
| | - Julia Gerbeth
- Department of Virology, Paul-Ehrlich-Institut, Paul-Ehrlich Strass 51-59, 63225 Langen, Germany; (F.D.H.); (L.H.); (C.v.R.); (J.G.)
| | - Imke Wieters
- Zentrum für Innere Medizin, Infektiologie, Universitätsklinikum Frankfurt, Goethe Universität Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (I.W.); (F.B.); (Y.K.); (M.J.G.T.V.)
| | - Frauke Borgans
- Zentrum für Innere Medizin, Infektiologie, Universitätsklinikum Frankfurt, Goethe Universität Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (I.W.); (F.B.); (Y.K.); (M.J.G.T.V.)
| | - Yascha Khodamoradi
- Zentrum für Innere Medizin, Infektiologie, Universitätsklinikum Frankfurt, Goethe Universität Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (I.W.); (F.B.); (Y.K.); (M.J.G.T.V.)
| | - Kai Zacharowski
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Frankfurt, Goethe Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany;
| | - Gernot Rohde
- Medizinische Klinik 1, Pneumologie/Allergologie, Universitätsklinikum Frankfurt, Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany;
| | - Maria J.G.T. Vehreschild
- Zentrum für Innere Medizin, Infektiologie, Universitätsklinikum Frankfurt, Goethe Universität Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (I.W.); (F.B.); (Y.K.); (M.J.G.T.V.)
| | - Barbara S. Schnierle
- Department of Virology, Paul-Ehrlich-Institut, Paul-Ehrlich Strass 51-59, 63225 Langen, Germany; (F.D.H.); (L.H.); (C.v.R.); (J.G.)
| |
Collapse
|
21
|
Vo HTM, Maestri A, Auerswald H, Sorn S, Lay S, Seng H, Sann S, Ya N, Pean P, Dussart P, Schwartz O, Ly S, Bruel T, Ly S, Duong V, Karlsson EA, Cantaert T. Robust and Functional Immune Memory Up to 9 Months After SARS-CoV-2 Infection: A Southeast Asian Longitudinal Cohort. Front Immunol 2022; 13:817905. [PMID: 35185909 PMCID: PMC8853741 DOI: 10.3389/fimmu.2022.817905] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/10/2022] [Indexed: 01/14/2023] Open
Abstract
The duration of humoral and cellular immune memory following SARS-CoV-2 infection in populations in least developed countries remains understudied but is key to overcome the current SARS-CoV-2 pandemic. Sixty-four Cambodian individuals with laboratory-confirmed infection with asymptomatic or mild/moderate clinical presentation were evaluated for Spike (S)-binding and neutralizing antibodies and antibody effector functions during acute phase of infection and at 6-9 months follow-up. Antigen-specific B cells, CD4+ and CD8+ T cells were characterized, and T cells were interrogated for functionality at late convalescence. Anti-S antibody titers decreased over time, but effector functions mediated by S-specific antibodies remained stable. S- and nucleocapsid (N)-specific B cells could be detected in late convalescence in the activated memory B cell compartment and are mostly IgG+. CD4+ and CD8+ T cell immune memory was maintained to S and membrane (M) protein. Asymptomatic infection resulted in decreased antibody-dependent cellular cytotoxicity (ADCC) and frequency of SARS-CoV-2-specific CD4+ T cells at late convalescence. Whereas anti-S antibodies correlated with S-specific B cells, there was no correlation between T cell response and humoral immune memory. Hence, all aspects of a protective immune response are maintained up to nine months after SARS-CoV-2 infection and in the absence of re-infection.
Collapse
Affiliation(s)
- Hoa Thi My Vo
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Alvino Maestri
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Heidi Auerswald
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sopheak Sorn
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sokchea Lay
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Heng Seng
- Department of Communicable Disease Control, Ministry of Health (CDC-MoH), Phnom Penh, Cambodia
| | - Sotheary Sann
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Nisa Ya
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Polidy Pean
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Philippe Dussart
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Olivier Schwartz
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France.,Vaccine Research Institute, Créteil, France
| | - Sovann Ly
- Department of Communicable Disease Control, Ministry of Health (CDC-MoH), Phnom Penh, Cambodia
| | - Timothée Bruel
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France.,Vaccine Research Institute, Créteil, France
| | - Sowath Ly
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Erik A Karlsson
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| |
Collapse
|
22
|
Natarajan H, Xu S, Crowley AR, Butler SE, Weiner JA, Bloch EM, Littlefield K, Benner SE, Shrestha R, Ajayi O, Wieland-Alter W, Sullivan D, Shoham S, Quinn TC, Casadevall A, Pekosz A, Redd AD, Tobian AAR, Connor RI, Wright PF, Ackerman ME. Antibody attributes that predict the neutralization and effector function of polyclonal responses to SARS-CoV-2. BMC Immunol 2022; 23:7. [PMID: 35172720 PMCID: PMC8851712 DOI: 10.1186/s12865-022-00480-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/07/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND While antibodies can provide significant protection from SARS-CoV-2 infection and disease sequelae, the specific attributes of the humoral response that contribute to immunity are incompletely defined. METHODS We employ machine learning to relate characteristics of the polyclonal antibody response raised by natural infection to diverse antibody effector functions and neutralization potency with the goal of generating both accurate predictions of each activity based on antibody response profiles as well as insights into antibody mechanisms of action. RESULTS To this end, antibody-mediated phagocytosis, cytotoxicity, complement deposition, and neutralization were accurately predicted from biophysical antibody profiles in both discovery and validation cohorts. These models identified SARS-CoV-2-specific IgM as a key predictor of neutralization activity whose mechanistic relevance was supported experimentally by depletion. CONCLUSIONS Validated models of how different aspects of the humoral response relate to antiviral antibody activities suggest desirable attributes to recapitulate by vaccination or other antibody-based interventions.
Collapse
Affiliation(s)
- Harini Natarajan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Shiwei Xu
- Program in Quantitative Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Andrew R Crowley
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Savannah E Butler
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Joshua A Weiner
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA
| | - Evan M Bloch
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kirsten Littlefield
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sarah E Benner
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ruchee Shrestha
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Olivia Ajayi
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Wendy Wieland-Alter
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - David Sullivan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Thomas C Quinn
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Arturo Casadevall
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew D Redd
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Aaron A R Tobian
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ruth I Connor
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Peter F Wright
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA.
- Program in Quantitative Biological Sciences, Dartmouth College, Hanover, NH, USA.
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA.
| |
Collapse
|
23
|
Ryan FJ, Hope CM, Masavuli MG, Lynn MA, Mekonnen ZA, Yeow AEL, Garcia-Valtanen P, Al-Delfi Z, Gummow J, Ferguson C, O'Connor S, Reddi BAJ, Hissaria P, Shaw D, Kok-Lim C, Gleadle JM, Beard MR, Barry SC, Grubor-Bauk B, Lynn DJ. Long-term perturbation of the peripheral immune system months after SARS-CoV-2 infection. BMC Med 2022; 20:26. [PMID: 35027067 PMCID: PMC8758383 DOI: 10.1186/s12916-021-02228-6] [Citation(s) in RCA: 171] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 12/29/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly infectious respiratory virus which is responsible for the coronavirus disease 2019 (COVID-19) pandemic. It is increasingly clear that recovered individuals, even those who had mild COVID-19, can suffer from persistent symptoms for many months after infection, a condition referred to as "long COVID", post-acute sequelae of COVID-19 (PASC), post-acute COVID-19 syndrome, or post COVID-19 condition. However, despite the plethora of research on COVID-19, relatively little is known about the molecular underpinnings of these long-term effects. METHODS We have undertaken an integrated analysis of immune responses in blood at a transcriptional, cellular, and serological level at 12, 16, and 24 weeks post-infection (wpi) in 69 patients recovering from mild, moderate, severe, or critical COVID-19 in comparison to healthy uninfected controls. Twenty-one of these patients were referred to a long COVID clinic and > 50% reported ongoing symptoms more than 6 months post-infection. RESULTS Anti-Spike and anti-RBD IgG responses were largely stable up to 24 wpi and correlated with disease severity. Deep immunophenotyping revealed significant differences in multiple innate (NK cells, LD neutrophils, CXCR3+ monocytes) and adaptive immune populations (T helper, T follicular helper, and regulatory T cells) in convalescent individuals compared to healthy controls, which were most strongly evident at 12 and 16 wpi. RNA sequencing revealed significant perturbations to gene expression in COVID-19 convalescents until at least 6 months post-infection. We also uncovered significant differences in the transcriptome at 24 wpi of convalescents who were referred to a long COVID clinic compared to those who were not. CONCLUSIONS Variation in the rate of recovery from infection at a cellular and transcriptional level may explain the persistence of symptoms associated with long COVID in some individuals.
Collapse
Affiliation(s)
- Feargal J Ryan
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, 5001, Australia
| | - Christopher M Hope
- Women's and Children's Health Network, North Adelaide, SA, Australia.,Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Makutiro G Masavuli
- Viral Immunology Group, Adelaide Medical School, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA, Australia
| | - Miriam A Lynn
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, 5001, Australia
| | - Zelalem A Mekonnen
- Viral Immunology Group, Adelaide Medical School, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA, Australia
| | - Arthur Eng Lip Yeow
- Viral Immunology Group, Adelaide Medical School, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA, Australia
| | - Pablo Garcia-Valtanen
- Viral Immunology Group, Adelaide Medical School, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA, Australia
| | - Zahraa Al-Delfi
- Viral Immunology Group, Adelaide Medical School, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA, Australia
| | - Jason Gummow
- Gene Silencing and Expression Core Facility, Adelaide Health and Medical Sciences, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Catherine Ferguson
- Infectious Diseases Department, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
| | - Stephanie O'Connor
- Intensive Care Unit, Royal Adelaide Hospital, Central Adelaide Local Health Network and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Benjamin A J Reddi
- Intensive Care Unit, Royal Adelaide Hospital, Central Adelaide Local Health Network and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Pravin Hissaria
- Infectious Diseases Department, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
| | - David Shaw
- Infectious Diseases Department, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
| | - Chuan Kok-Lim
- Infectious Diseases Department, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia.,Microbiology and Infectious Diseases Department, SA Pathology, Adelaide, SA, Australia
| | - Jonathan M Gleadle
- Department of Renal Medicine, Flinders Medical Centre, Flinders University, Bedford Park, SA, 5042, Australia.,Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, 5042, Australia
| | - Michael R Beard
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Simon C Barry
- Women's and Children's Health Network, North Adelaide, SA, Australia. .,Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia.
| | - Branka Grubor-Bauk
- Viral Immunology Group, Adelaide Medical School, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA, Australia.
| | - David J Lynn
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, 5001, Australia. .,Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, 5042, Australia.
| |
Collapse
|
24
|
Soeorg H, Jõgi P, Naaber P, Ottas A, Toompere K, Lutsar I. Seroprevalence and levels of IgG antibodies after COVID-19 infection or vaccination. Infect Dis (Lond) 2022; 54:63-71. [PMID: 34520315 PMCID: PMC8442755 DOI: 10.1080/23744235.2021.1974540] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/19/2021] [Accepted: 08/25/2021] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND In a country-wide seroprevalence study of COVID-19 in Estonia, we aimed to determine the seroprevalence and the dynamics of IgG against SARS-CoV-2 after vaccination or positive PCR-test. METHODS Leftover blood samples were selected between 8 February and 25 March 2021, by SYNLAB Estonia from all counties and age groups (0-9, 10-19, 20-59, 60-69, 70-79 and 80-100 years) proportionally to the whole Estonian population and tested for IgG against SARS-CoV-2 spike protein receptor-binding domain (anti-S-RBD IgG) using Abbott SARS-CoV-2 IgG II Quant assay. Antibody levels after positive PCR-test or vaccination were described by exponential increase-decrease models. RESULTS According to total of 2517 samples, overall seroprevalence (95% confidence interval [CI]) was 20.1% (18.5-21.7%), similar in all age groups, but varied between counties. If individuals vaccinated with the first dose at least 14 d before antibody measurement were assumed to be seronegative, the overall seroprevalence was 15.8% (14.4-17.3%), 4.0-fold larger than the proportion of PCR-confirmed COVID-19 cases. Of seropositive individuals (n = 506) 194 (38.3%; 33.8-43.1%) had not had positive PCR-test or been vaccinated. According to exponential increase-decrease model, the peak of anti-S-RBD IgG in a 52-year-old (median age of PCR-positive and/or vaccinated individuals) was significantly higher after vaccination compared with positive PCR-test (22,082 (12,897-26,875) vs. 6732 (2321-8243) AU/mL), but half-life was similar (26.5 (6.9-46.1) vs. 38.3 (8.2-68.5) d). CONCLUSIONS One year after the start of COVID-19 pandemic the actual prevalence of infection is still underestimated compared with PCR-confirmed COVID-19 cases. Older compared with younger individuals have lower anti-S-RBD IgG level after vaccination, but similar decline rate.
Collapse
Affiliation(s)
- Hiie Soeorg
- Department of Microbiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Piia Jõgi
- Children’s Clinic of Tartu University Hospital, Tartu, Estonia
- Department of Pediatrics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Paul Naaber
- Department of Microbiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- SYNLAB Estonia, Veerenni 53a, Tallinn, Estonia
| | - Aigar Ottas
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Karolin Toompere
- Department of Epidemiology and Biostatistics, Institute of Family Medicine and Public Health, University of Tartu, Tartu, Estonia
| | - Irja Lutsar
- Department of Microbiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
25
|
Amanat F, Strohmeier S, Meade PS, Dambrauskas N, Mühlemann B, Smith DJ, Vigdorovich V, Sather DN, Coughlan L, Krammer F. Vaccination with SARS-CoV-2 variants of concern protects mice from challenge with wild-type virus. PLoS Biol 2021; 19:e3001384. [PMID: 34914685 PMCID: PMC8758087 DOI: 10.1371/journal.pbio.3001384] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 01/13/2022] [Accepted: 11/24/2021] [Indexed: 12/23/2022] Open
Abstract
Vaccines against Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) have been highly efficient in protecting against Coronavirus Disease 2019 (COVID-19). However, the emergence of viral variants that are more transmissible and, in some cases, escape from neutralizing antibody responses has raised concerns. Here, we evaluated recombinant protein spike antigens derived from wild-type SARS-CoV-2 and from variants B.1.1.7, B.1.351, and P.1 for their immunogenicity and protective effect in vivo against challenge with wild-type SARS-CoV-2 in the mouse model. All proteins induced high neutralizing antibodies against the respective viruses but also induced high cross-neutralizing antibody responses. The decline in neutralizing titers between variants was moderate, with B.1.1.7-vaccinated animals having a maximum fold reduction of 4.8 against B.1.351 virus. P.1 induced the most cross-reactive antibody responses but was also the least immunogenic in terms of homologous neutralization titers. However, all antigens protected from challenge with wild-type SARS-CoV-2 in a mouse model.
Collapse
Affiliation(s)
- Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Philip S. Meade
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Nicholas Dambrauskas
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Barbara Mühlemann
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Centre for Infection Research (DZIF), partner site Charité, Berlin, Germany
| | - Derek J. Smith
- Centre for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - Vladimir Vigdorovich
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - D. Noah Sather
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Lynda Coughlan
- University of Maryland School of Medicine, Department of Microbiology and Immunology, Baltimore, Maryland, United States of America
- University of Maryland School of Medicine, Center for Vaccine Development and Global Health (CVD), Baltimore, Maryland, United States of America
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
26
|
Jiang Y, Duffy F, Hadlock J, Raappana A, Styrchak S, Beck I, Mast FD, Miller LR, Chour W, Houck J, Armistead B, Duvvuri VR, Yeung W, Haglund M, Wallner J, Wallick JA, Hardy S, Oldroyd A, Ko D, Gervassi A, Murray KM, Kaplan H, Aitchison JD, Heath JR, Sather DN, Goldman JD, Frenkel L, Harrington WE. Angiotensin II receptor I auto-antibodies following SARS-CoV-2 infection. PLoS One 2021; 16:e0259902. [PMID: 34788328 PMCID: PMC8598062 DOI: 10.1371/journal.pone.0259902] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/28/2021] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is associated with endothelial activation and coagulopathy, which may be related to pre-existing or infection-induced pro-thrombotic autoantibodies such as those targeting angiotensin II type I receptor (AT1R-Ab). METHODS We compared prevalence and levels of AT1R-Ab in COVID-19 cases with mild or severe disease to age and sex matched negative controls utilizing multivariate logistic and quantile regression adjusted for comorbidities including hypertension, diabetes, and heart disease. RESULTS There were trends toward increased prevalence (50% vs. 33%, p = 0.1) and level of AT1R-Ab (median 9.8 vs. 6.1 U/mL, p = 0.06) in all cases versus controls. When considered by COVID-19 disease severity, there was a trend toward increased prevalence of AT1R-Ab (55% vs. 31%, p = 0.07), as well as significantly higher AT1R-Ab levels (median 10.7 vs. 5.9 U/mL, p = 0.03) amongst individuals with mild COVID-19 versus matched controls. In contrast, the prevalence (42% vs. 37%, p = 0.9) and level (both medians 6.7 U/mL, p = 0.9) of AT1R-Ab amongst those with severe COVID-19 did not differ from matched controls. CONCLUSIONS These findings support an association between COVID-19 and AT1R-Ab, emphasizing that vascular pathology may be present in individuals with mild COVID-19 as well as those with severe disease.
Collapse
Affiliation(s)
- Yonghou Jiang
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Fergal Duffy
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Jennifer Hadlock
- Institute for Systems Biology, Seattle, Washington, United States of America
- Providence St. Joseph Health, Renton, Washington, United States of America
| | - Andrew Raappana
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Sheila Styrchak
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Ingrid Beck
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Fred D. Mast
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Leslie R. Miller
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - William Chour
- Institute for Systems Biology, Seattle, Washington, United States of America
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - John Houck
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Blair Armistead
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Venkata R. Duvvuri
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Winnie Yeung
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Micaela Haglund
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Jackson Wallner
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Julie A. Wallick
- Providence St. Joseph Health, Renton, Washington, United States of America
- Swedish Center for Research and Innovation, Swedish Medical Center, Seattle, Washington, United States of America
| | - Samantha Hardy
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Alyssa Oldroyd
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Daisy Ko
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Ana Gervassi
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Kim M. Murray
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Henry Kaplan
- Swedish Cancer Institute, Swedish Medical Center, Seattle, Washington, United States of America
| | - John D. Aitchison
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - James R. Heath
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - D. Noah Sather
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Jason D. Goldman
- Providence St. Joseph Health, Renton, Washington, United States of America
- Swedish Center for Research and Innovation, Swedish Medical Center, Seattle, Washington, United States of America
- Division of Allergy & Infectious Diseases, University of Washington, Seattle, Washington, United States of America
| | - Lisa Frenkel
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Whitney E. Harrington
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
27
|
Al-Karmalawy AA, Soltane R, Abo Elmaaty A, Tantawy MA, Antar SA, Yahya G, Chrouda A, Pashameah RA, Mustafa M, Abu Mraheil M, Mostafa A. Coronavirus Disease (COVID-19) Control between Drug Repurposing and Vaccination: A Comprehensive Overview. Vaccines (Basel) 2021; 9:1317. [PMID: 34835248 PMCID: PMC8622998 DOI: 10.3390/vaccines9111317] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023] Open
Abstract
Respiratory viruses represent a major public health concern, as they are highly mutated, resulting in new strains emerging with high pathogenicity. Currently, the world is suffering from the newly evolving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This virus is the cause of coronavirus disease 2019 (COVID-19), a mild-to-severe respiratory tract infection with frequent ability to give rise to fatal pneumonia in humans. The overwhelming outbreak of SARS-CoV-2 continues to unfold all over the world, urging scientists to put an end to this global pandemic through biological and pharmaceutical interventions. Currently, there is no specific treatment option that is capable of COVID-19 pandemic eradication, so several repurposed drugs and newly conditionally approved vaccines are in use and heavily applied to control the COVID-19 pandemic. The emergence of new variants of the virus that partially or totally escape from the immune response elicited by the approved vaccines requires continuous monitoring of the emerging variants to update the content of the developed vaccines or modify them totally to match the new variants. Herein, we discuss the potential therapeutic and prophylactic interventions including repurposed drugs and the newly developed/approved vaccines, highlighting the impact of virus evolution on the immune evasion of the virus from currently licensed vaccines for COVID-19.
Collapse
Affiliation(s)
- Ahmed A. Al-Karmalawy
- Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Raya Soltane
- Department of Basic Sciences, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (R.S.); (R.A.P.)
- Department of Biology, Faculty of Sciences, Tunis El Manar University, Tunis 1068, Tunisia
| | - Ayman Abo Elmaaty
- Department of Medicinal Chemistry, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt;
| | - Mohamed A. Tantawy
- Hormones Department, Medical Research and Clinical Studies Research Institute, National Research Centre, Dokki 12622, Egypt;
- Stem Cells Laboratory, Center of Excellence for Advanced Sciences, National Research Centre, Dokki 12622, Egypt
| | - Samar A. Antar
- Department of Pharmacology, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt;
| | - Galal Yahya
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - Amani Chrouda
- Department of Chemistry, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11932, Saudi Arabia;
- Laboratory of Interfaces and Advanced Materials, Faculty of Sciences, Monastir University, Monastir 5000, Tunisia
- Institute of Analytical Sciences, UMR CNRS-UCBL-ENS 5280, 5 Rue la Doua, CEDEX, 69100 Villeurbanne, France
| | - Rami Adel Pashameah
- Department of Basic Sciences, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (R.S.); (R.A.P.)
| | - Muhamad Mustafa
- Department of Medicinal Chemistry, Deraya University, Minia 61111, Egypt;
| | - Mobarak Abu Mraheil
- German Center for Infection Research (DZIF), Institute of Medical Microbiology, Justus-Liebig University, 35392 Giessen, Germany;
| | - Ahmed Mostafa
- German Center for Infection Research (DZIF), Institute of Medical Microbiology, Justus-Liebig University, 35392 Giessen, Germany;
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Dokki 12622, Egypt
| |
Collapse
|
28
|
Cappuccilli M, Bruno PF, Spazzoli A, Righini M, Flachi M, Semprini S, Grumiro L, Marino MM, Schiavone P, Fabbri E, Fantini M, Buscaroli A, Rigotti A, La Manna G, Sambri V, Mosconi G. Persistence of Antibody Responses to the SARS-CoV-2 in Dialysis Patients and Renal Transplant Recipients Recovered from COVID-19. Pathogens 2021; 10:1289. [PMID: 34684237 PMCID: PMC8541005 DOI: 10.3390/pathogens10101289] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/27/2021] [Accepted: 10/04/2021] [Indexed: 12/14/2022] Open
Abstract
Nephropathic subjects with impaired immune responses show dramatically high infection rates of coronavirus disease 2019 (COVID-19). This work evaluated the ability to acquire and maintain protective antibodies over time in 26 hemodialysis patients and 21 kidney transplant recipients. The subjects were followed-up through quantitative determination of circulating SARS-CoV-2 S1/S2 IgG and neutralizing antibodies in the 6-month period after clinical and laboratory recovery. A group of 143 healthcare workers with no underlying chronic pathologies or renal diseases recovered from COVID was also evaluated. In both dialysis and transplanted patients, antibody titers reached a zenith around the 3rd month, and then a decline occurred on average between the 270th and 300th day. Immunocompromised patients who lost antibodies around the 6th month were more common than non-renal subjects, although the difference was not significant (38.5% vs. 26.6%). Considering the decay of antibody levels below the positivity threshold (15 AU/mL) as "failure", a progressive loss of immunisation was found in the overall population starting 6 months after recovery. A longer overall antibody persistence was observed in severe forms of COVID-19 (p = 0.0183), but within each group, given the small number of patients, the difference was not significant (dialysis: p = 0.0702; transplant: p = 0.1899). These data suggest that immunocompromised renal patients recovered from COVID-19 have weakened and heterogeneous humoral responses that tend to decay over time. Despite interindividual variability, an association emerged between antibody persistence and clinical severity, similar to the subjects with preserved immune function.
Collapse
Affiliation(s)
- Maria Cappuccilli
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (M.C.); (G.L.M.)
| | - Paolo Ferdinando Bruno
- Nephrology and Dialysis Unit, AUSL Romagna Morgagni-Pierantoni Hospital, 47121 Forlì, Italy; (P.F.B.); (A.S.)
| | - Alessandra Spazzoli
- Nephrology and Dialysis Unit, AUSL Romagna Morgagni-Pierantoni Hospital, 47121 Forlì, Italy; (P.F.B.); (A.S.)
| | - Matteo Righini
- Nephrology and Dialysis Unit, AUSL Romagna S. Maria delle Croci Hospital, 48121 Ravenna, Italy; (M.R.); (A.B.)
| | - Marta Flachi
- Nephrology and Dialysis Unit, AUSL Romagna Infermi Hospital, 47923 Rimini, Italy; (M.F.); (A.R.)
| | - Simona Semprini
- Unit of Microbiology, AUSL Romagna Laboratory, 47023 Pievesestina, Italy; (S.S.); (L.G.); (M.M.M.); (P.S.); (V.S.)
| | - Laura Grumiro
- Unit of Microbiology, AUSL Romagna Laboratory, 47023 Pievesestina, Italy; (S.S.); (L.G.); (M.M.M.); (P.S.); (V.S.)
| | - Maria Michela Marino
- Unit of Microbiology, AUSL Romagna Laboratory, 47023 Pievesestina, Italy; (S.S.); (L.G.); (M.M.M.); (P.S.); (V.S.)
| | - Pasqua Schiavone
- Unit of Microbiology, AUSL Romagna Laboratory, 47023 Pievesestina, Italy; (S.S.); (L.G.); (M.M.M.); (P.S.); (V.S.)
| | - Elisabetta Fabbri
- Local Healthcare Authority of Romagna (AUSL Romagna), 48121 Ravenna, Italy; (E.F.); (M.F.)
| | - Michela Fantini
- Local Healthcare Authority of Romagna (AUSL Romagna), 48121 Ravenna, Italy; (E.F.); (M.F.)
| | - Andrea Buscaroli
- Nephrology and Dialysis Unit, AUSL Romagna S. Maria delle Croci Hospital, 48121 Ravenna, Italy; (M.R.); (A.B.)
| | - Angelo Rigotti
- Nephrology and Dialysis Unit, AUSL Romagna Infermi Hospital, 47923 Rimini, Italy; (M.F.); (A.R.)
| | - Gaetano La Manna
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (M.C.); (G.L.M.)
| | - Vittorio Sambri
- Unit of Microbiology, AUSL Romagna Laboratory, 47023 Pievesestina, Italy; (S.S.); (L.G.); (M.M.M.); (P.S.); (V.S.)
| | - Giovanni Mosconi
- Nephrology and Dialysis Unit, AUSL Romagna Morgagni-Pierantoni Hospital, 47121 Forlì, Italy; (P.F.B.); (A.S.)
| |
Collapse
|
29
|
Garrido C, Hurst JH, Lorang CG, Aquino JN, Rodriguez J, Pfeiffer TS, Singh T, Semmes EC, Lugo DJ, Rotta AT, Turner NA, Burke TW, McClain MT, Petzold EA, Permar SR, Moody MA, Woods CW, Kelly MS, Fouda GG. Asymptomatic or mild symptomatic SARS-CoV-2 infection elicits durable neutralizing antibody responses in children and adolescents. JCI Insight 2021; 6:150909. [PMID: 34228642 PMCID: PMC8492306 DOI: 10.1172/jci.insight.150909] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/30/2021] [Indexed: 11/17/2022] Open
Abstract
As SARS-CoV-2 continues to spread globally, questions have emerged regarding the strength and durability of immune responses in specific populations. In this study, we evaluated humoral immune responses in 69 children and adolescents with asymptomatic or mild symptomatic SARS-CoV-2 infection. We detected robust IgM, IgG, and IgA antibody responses to a broad array of SARS-CoV-2 antigens at the time of acute infection and 2 and 4 months after acute infection in all participants. Notably, these antibody responses were associated with virus-neutralizing activity that was still detectable 4 months after acute infection in 94% of children. Moreover, antibody responses and neutralizing activity in sera from children and adolescents were comparable or superior to those observed in sera from 24 adults with mild symptomatic infection. Taken together, these findings indicate that children and adolescents with mild or asymptomatic SARS-CoV-2 infection generate robust and durable humoral immune responses that can likely contribute to protection from reinfection.
Collapse
Affiliation(s)
| | - Jillian H. Hurst
- Department of Pediatrics, Division of Infectious Diseases
- Children’s Health & Discovery Institute, Department of Pediatrics, and
| | | | | | - Javier Rodriguez
- Children’s Clinical Research Unit, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | | | | | - Eleanor C. Semmes
- Duke Human Vaccine Institute
- Children’s Health & Discovery Institute, Department of Pediatrics, and
- Medical Scientist Training Program, Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Debra J. Lugo
- Department of Pediatrics, Division of Infectious Diseases
| | - Alexandre T. Rotta
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, and
| | - Nicholas A. Turner
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, USA
| | - Thomas W. Burke
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, North Carolina, USA
| | - Micah T. McClain
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, USA
- Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, North Carolina, USA
| | - Elizabeth A. Petzold
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, North Carolina, USA
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell School of Medicine, New York City, New York, USA
| | - M. Anthony Moody
- Duke Human Vaccine Institute
- Department of Pediatrics, Division of Infectious Diseases
| | - Christopher W. Woods
- Duke Human Vaccine Institute
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, USA
- Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, North Carolina, USA
| | | | - Genevieve G. Fouda
- Duke Human Vaccine Institute
- Department of Pediatrics, Division of Infectious Diseases
| |
Collapse
|
30
|
Kumavath R, Barh D, Andrade BS, Imchen M, Aburjaile FF, Ch A, Rodrigues DLN, Tiwari S, Alzahrani KJ, Góes-Neto A, Weener ME, Ghosh P, Azevedo V. The Spike of SARS-CoV-2: Uniqueness and Applications. Front Immunol 2021; 12:663912. [PMID: 34305894 PMCID: PMC8297464 DOI: 10.3389/fimmu.2021.663912] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/16/2021] [Indexed: 12/20/2022] Open
Abstract
The Spike (S) protein of the SARS-CoV-2 virus is critical for its ability to attach and fuse into the host cells, leading to infection, and transmission. In this review, we have initially performed a meta-analysis of keywords associated with the S protein to frame the outline of important research findings and directions related to it. Based on this outline, we have reviewed the structure, uniqueness, and origin of the S protein of SARS-CoV-2. Furthermore, the interactions of the Spike protein with host and its implications in COVID-19 pathogenesis, as well as drug and vaccine development, are discussed. We have also summarized the recent advances in detection methods using S protein-based RT-PCR, ELISA, point-of-care lateral flow immunoassay, and graphene-based field-effect transistor (FET) biosensors. Finally, we have also discussed the emerging Spike mutants and the efficacy of the Spike-based vaccines against those strains. Overall, we have covered most of the recent advances on the SARS-CoV-2 Spike protein and its possible implications in countering this virus.
Collapse
Affiliation(s)
- Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kasaragod, India
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal, India
- Laboratório de Genética Celular e Molecular, Departamento de Genetica, Ecologia e Evolucao, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bruno Silva Andrade
- Laboratório de Bioinformática e Química Computacional, Departamento de Ciências Biológicas, Universidade Estadual do Sudoeste da Bahia (UESB), Jequié, Brazil
| | - Madangchanok Imchen
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kasaragod, India
| | - Flavia Figueira Aburjaile
- Laboratório de Genética Celular e Molecular, Departamento de Genetica, Ecologia e Evolucao, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Athira Ch
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kasaragod, India
| | - Diego Lucas Neres Rodrigues
- Laboratório de Genética Celular e Molecular, Departamento de Genetica, Ecologia e Evolucao, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sandeep Tiwari
- Laboratório de Genética Celular e Molecular, Departamento de Genetica, Ecologia e Evolucao, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Aristóteles Góes-Neto
- Laboratório de Biologia Molecular e Computacional de Fungos, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | | | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA, United States
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular, Departamento de Genetica, Ecologia e Evolucao, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
31
|
Iruretagoyena M, Vial MR, Spencer-Sandino M, Gaete P, Peters A, Delgado I, Perez I, Calderon C, Porte L, Legarraga P, Anderson A, Aguilera X, Vial P, Weitzel T, Munita JM. Longitudinal assessment of SARS-CoV-2 IgG seroconversionamong front-line healthcare workers during the first wave of the Covid-19 pandemic at a tertiary-care hospital in Chile. BMC Infect Dis 2021; 21:478. [PMID: 34039287 PMCID: PMC8149923 DOI: 10.1186/s12879-021-06208-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/12/2021] [Indexed: 12/18/2022] Open
Abstract
Background Healthcare workers (HCWs) are at high risk of exposure to SARS-CoV-2. Cross-sectional studies have provided variable rates of seroprevalence in HCWs. Longitudinal assessments of the serological response to Covid-19 among HCWs are crucial to understanding the risk of infection and changes in antibody titers over time. We aimed to investigate seroprevalence and risk factors associated with seroconversion in a prospective cohort of HCWs during the peak of the first wave of the Covid-19 pandemic. Methods We conducted a longitudinal study among 446 front-line HCWsin a tertiary-care hospital in Chile from April to July 2020. IgG was determined monthly using two different ELISAs in serum samples of HCWs, during the three-month period. In each visit, demographic data, symptoms, risk factors, and exposure risks were also assessed. Results The overall seroprevalence at the end of the study period was 24% (95% CI20.2–28.3), with 43% of seropositive HCWs reporting no prior symptoms. Seroconversion rates significantly differed over the study period, from 2.1% to as high as 8.8% at the peak of the epidemic. There were no statistically significant differences observed between HCWs in direct clinical care of patients with Covid-19 and those working in low risk areas. Antibody titers appeared to wane over time. Conclusions HCWs were severely affected with a high rate of seroconversion that appeared to mirror the local epidemiological situation. A significant amount of participants underwent an asymptomatic infection, highlighting the need for improved surveillance policies. Antibody titers appear to wane over time; further studies to understand this finding’s impact on the risk of reinfection are warranted. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06208-2.
Collapse
Affiliation(s)
- Mirentxu Iruretagoyena
- Laboratorio Clínico, Clínica Alemana de Santiago, Santiago, Chile.,Facultad de Medicina Clínica Alemana, Universidad del Desarrollo (CAS-UDD), Santiago, Chile
| | - Macarena R Vial
- Facultad de Medicina Clínica Alemana, Universidad del Desarrollo (CAS-UDD), Santiago, Chile
| | - Maria Spencer-Sandino
- Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina CAS-UDD, Santiago, Chile.,Millennium Initiative for Collaborative Research on Bacterial Resistance (MICROB-R), Santiago, Chile
| | - Pablo Gaete
- Facultad de Medicina Clínica Alemana, Universidad del Desarrollo (CAS-UDD), Santiago, Chile
| | - Anne Peters
- Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina CAS-UDD, Santiago, Chile.,Millennium Initiative for Collaborative Research on Bacterial Resistance (MICROB-R), Santiago, Chile
| | - Iris Delgado
- Facultad de Medicina Clínica Alemana, Universidad del Desarrollo (CAS-UDD), Santiago, Chile.,Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina CAS-UDD, Santiago, Chile
| | - Inia Perez
- Facultad de Medicina Clínica Alemana, Universidad del Desarrollo (CAS-UDD), Santiago, Chile
| | - Claudia Calderon
- Facultad de Medicina Clínica Alemana, Universidad del Desarrollo (CAS-UDD), Santiago, Chile
| | - Lorena Porte
- Laboratorio Clínico, Clínica Alemana de Santiago, Santiago, Chile.,Facultad de Medicina Clínica Alemana, Universidad del Desarrollo (CAS-UDD), Santiago, Chile
| | - Paulette Legarraga
- Laboratorio Clínico, Clínica Alemana de Santiago, Santiago, Chile.,Facultad de Medicina Clínica Alemana, Universidad del Desarrollo (CAS-UDD), Santiago, Chile
| | - Alicia Anderson
- Laboratorio Clínico, Clínica Alemana de Santiago, Santiago, Chile.,Facultad de Medicina Clínica Alemana, Universidad del Desarrollo (CAS-UDD), Santiago, Chile
| | - Ximena Aguilera
- Facultad de Medicina Clínica Alemana, Universidad del Desarrollo (CAS-UDD), Santiago, Chile.,Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina CAS-UDD, Santiago, Chile
| | - Pablo Vial
- Facultad de Medicina Clínica Alemana, Universidad del Desarrollo (CAS-UDD), Santiago, Chile.,Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina CAS-UDD, Santiago, Chile
| | - Thomas Weitzel
- Laboratorio Clínico, Clínica Alemana de Santiago, Santiago, Chile.,Facultad de Medicina Clínica Alemana, Universidad del Desarrollo (CAS-UDD), Santiago, Chile.,Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina CAS-UDD, Santiago, Chile
| | - Jose M Munita
- Facultad de Medicina Clínica Alemana, Universidad del Desarrollo (CAS-UDD), Santiago, Chile. .,Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina CAS-UDD, Santiago, Chile. .,Millennium Initiative for Collaborative Research on Bacterial Resistance (MICROB-R), Santiago, Chile.
| |
Collapse
|