1
|
Edwards CT, Karunakaran KA, Garcia E, Beutler N, Gagne M, Golden N, Aoued H, Pellegrini KL, Burnett MR, Honeycutt CC, Lapp SA, Ton T, Lin MC, Metz A, Bombin A, Goff K, Scheuermann SE, Wilkes A, Wood JS, Ehnert S, Weissman S, Curran EH, Roy M, Dessasau E, Paiardini M, Upadhyay AA, Moore IN, Maness NJ, Douek DC, Piantadosi A, Andrabi R, Rogers TR, Burton DR, Bosinger SE. Passive infusion of an S2-Stem broadly neutralizing antibody protects against SARS-CoV-2 infection and lower airway inflammation in rhesus macaques. PLoS Pathog 2025; 21:e1012456. [PMID: 39847599 PMCID: PMC11793774 DOI: 10.1371/journal.ppat.1012456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 02/04/2025] [Accepted: 12/27/2024] [Indexed: 01/25/2025] Open
Abstract
The continued evolution of SARS-CoV-2 variants capable of subverting vaccine and infection-induced immunity suggests the advantage of a broadly protective vaccine against betacoronaviruses (β-CoVs). Recent studies have isolated monoclonal antibodies (mAbs) from SARS-CoV-2 recovered-vaccinated donors capable of neutralizing many variants of SARS-CoV-2 and other β-CoVs. Many of these mAbs target the conserved S2 stem region of the SARS-CoV-2 spike protein, rather than the receptor binding domain contained within S1 primarily targeted by current SARS-CoV-2 vaccines. One of these S2-directed mAbs, CC40.8, has demonstrated protective efficacy in small animal models against SARS-CoV-2 challenge. As the next step in the pre-clinical testing of S2-directed antibodies as a strategy to protect from SARS-CoV-2 infection, we evaluated the in vivo efficacy of CC40.8 in a clinically relevant non-human primate model by conducting passive antibody transfer to rhesus macaques (RM) followed by SARS-CoV-2 challenge. CC40.8 mAb was intravenously infused at 10mg/kg, 1mg/kg, or 0.1 mg/kg into groups (n = 6) of RM, alongside one group that received a control antibody (PGT121). Viral loads in the lower airway were significantly reduced in animals receiving higher doses of CC40.8. We observed a significant reduction in inflammatory cytokines and macrophages within the lower airway of animals infused with 10mg/kg and 1mg/kg doses of CC40.8. Viral genome sequencing demonstrated a lack of escape mutations in the CC40.8 epitope. Collectively, these data demonstrate the protective efficiency of broadly neutralizing S2-targeting antibodies against SARS-CoV-2 infection within the lower airway while providing critical preclinical work necessary for the development of pan-β-CoV vaccines.
Collapse
Affiliation(s)
- Christopher T. Edwards
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Kirti A. Karunakaran
- Department of Pathology, Microbiology & Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States of America
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Matthew Gagne
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nadia Golden
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Hadj Aoued
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Kathryn L. Pellegrini
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Matthew R. Burnett
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Christopher Cole Honeycutt
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Stacey A. Lapp
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Thang Ton
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Mark C. Lin
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Amanda Metz
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Andrei Bombin
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kelly Goff
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Sarah E. Scheuermann
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Amelia Wilkes
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Jennifer S. Wood
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Stephanie Ehnert
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Stacey Weissman
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Elizabeth H. Curran
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Melissa Roy
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Evan Dessasau
- Division of Histology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Amit A. Upadhyay
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Ian N. Moore
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Nicholas J. Maness
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Daniel C. Douek
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anne Piantadosi
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, California, United States of America
| | - Thomas R. Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, California, United States of America
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, United States of America
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
2
|
Lee P, Kim J, Oh H, Kim CU, Jeong AY, Lee MS, Jang MS, Hong JJ, Park JE, Kim DJ. Coronavirus nucleocapsid-based vaccine provides partial protection against hetero-species coronavirus in murine models. Antiviral Res 2024; 231:105991. [PMID: 39181216 DOI: 10.1016/j.antiviral.2024.105991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/06/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Most coronavirus vaccines focus on the spike (S) antigen, but the frequent mutations in S raise concerns about the vaccine efficacy against new variants. Although additional antigens with conserved sequences are have been tested, the extent to which these vaccines can provide immunity against different coronavirus species remains unclear. In this study, we assessed the potential of nucleocapsid (N) as a coronavirus vaccine antigen. Immunization with MERS-CoV N induced robust immune responses, providing significant protection against MERS-CoV. Notably, MERS-CoV N elicited cross-reactive T cell responses to SARS-CoV-2 N and significantly reduced lung inflammation following a SARS-CoV-2 challenge in the transient hACE2 mouse model. However, in K18-hACE transgenic mice, the vaccine showed limited protection. Collectively, our findings suggest that coronavirus N can be an effective vaccine antigen against homologous viruses, but its efficacy may vary across different coronaviruses, highlighting the need for further research on pan-coronavirus vaccines using conserved antigens.
Collapse
Affiliation(s)
- Pureum Lee
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; University of Science and Technology (UST), Daejeon, South Korea
| | - Jihee Kim
- Chungnam National University College of Veterinary Medicine, Daejeon, South Korea
| | - Hanseul Oh
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; Chungbuk National University College of Veterinary Medicine, Cheongju, South Korea
| | - Chang-Ung Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Ahn Young Jeong
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; Princeton University, Princeton, NJ, USA
| | - Moo-Seung Lee
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; University of Science and Technology (UST), Daejeon, South Korea
| | | | - Jung Joo Hong
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.
| | - Jung-Eun Park
- Chungnam National University College of Veterinary Medicine, Daejeon, South Korea.
| | - Doo-Jin Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; Chungbuk National University College of Medicine, Cheongju, South Korea; Biomedical Research Institute, Chungbuk National University Hospital, Cheongju, South Korea.
| |
Collapse
|
3
|
Edwards CT, Karunakaran KA, Garcia E, Beutler N, Gagne M, Golden N, Aoued H, Pellegrini KL, Burnett MR, Honeycutt CC, Lapp SA, Ton T, Lin MC, Metz A, Bombin A, Goff K, Scheuermann SE, Wilkes A, Wood JS, Ehnert S, Weissman S, Curran EH, Roy M, Dessasau E, Paiardini M, Upadhyay AA, Moore I, Maness NJ, Douek DC, Piantadosi A, Andrabi R, Rogers TR, Burton DR, Bosinger SE. Passive infusion of an S2-Stem broadly neutralizing antibody protects against SARS-CoV-2 infection and lower airway inflammation in rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605768. [PMID: 39109178 PMCID: PMC11302620 DOI: 10.1101/2024.07.30.605768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The continued evolution of SARS-CoV-2 variants capable of subverting vaccine and infection-induced immunity suggests the advantage of a broadly protective vaccine against betacoronaviruses (β-CoVs). Recent studies have isolated monoclonal antibodies (mAbs) from SARS-CoV-2 recovered-vaccinated donors capable of neutralizing many variants of SARS-CoV-2 and other β-CoVs. Many of these mAbs target the conserved S2 stem region of the SARS-CoV-2 spike protein, rather the receptor binding domain contained within S1 primarily targeted by current SARS-CoV-2 vaccines. One of these S2-directed mAbs, CC40.8, has demonstrated protective efficacy in small animal models against SARS-CoV-2 challenge. As the next step in the pre-clinical testing of S2-directed antibodies as a strategy to protect from SARS-CoV-2 infection, we evaluated the in vivo efficacy of CC40.8 in a clinically relevant non-human primate model by conducting passive antibody transfer to rhesus macaques (RM) followed by SARS-CoV-2 challenge. CC40.8 mAb was intravenously infused at 10mg/kg, 1mg/kg, or 0.1 mg/kg into groups (n=6) of RM, alongside one group that received a control antibody (PGT121). Viral loads in the lower airway were significantly reduced in animals receiving higher doses of CC40.8. We observed a significant reduction in inflammatory cytokines and macrophages within the lower airway of animals infused with 10mg/kg and 1mg/kg doses of CC40.8. Viral genome sequencing demonstrated a lack of escape mutations in the CC40.8 epitope. Collectively, these data demonstrate the protective efficiency of broadly neutralizing S2-targeting antibodies against SARS-CoV-2 infection within the lower airway while providing critical preclinical work necessary for the development of pan-β-CoV vaccines.
Collapse
Affiliation(s)
- Christopher T. Edwards
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Kirti A. Karunakaran
- Department of Pathology, Microbiology & Immunology, Vanderbilt University, Nashville, TN 37235, USA
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, Minnesota 55356, USA
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Matthew Gagne
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nadia Golden
- Tulane National Primate Research Center, Covington, LA, USA
| | - Hadj Aoued
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Kathryn L. Pellegrini
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Matthew R. Burnett
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Cole Honeycutt
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stacey A. Lapp
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Thang Ton
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Mark C. Lin
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Amanda Metz
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Andrei Bombin
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Kelly Goff
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Amelia Wilkes
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jennifer S. Wood
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Stephanie Ehnert
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Stacey Weissman
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Elizabeth H. Curran
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Melissa Roy
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Evan Dessasau
- Division of Histology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Amit A. Upadhyay
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Ian Moore
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Daniel C. Douek
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anne Piantadosi
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Thomas R. Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139, USA
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
4
|
Berguido FJ, Kangethe RT, Shell W, Wijewardana V, Grabherr R, Cattoli G, Lamien CE. Different Neutralizing Antibody Responses of Heterologous Sera on Sheeppox and Lumpy Skin Disease Viruses. Viruses 2024; 16:1127. [PMID: 39066289 PMCID: PMC11281510 DOI: 10.3390/v16071127] [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: 06/12/2024] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Sheeppox virus (SPPV), goatpox virus (GTPV), and lumpy skin disease virus (LSDV) are the three members of the genus Capripoxvirus within the Poxviridae family and are the etiologic agents of sheeppox (SPP), goatpox (GTP), and lumpy skin disease (LSD), respectively. LSD, GTP, and SPP are endemic in Africa and Asia, causing severe disease outbreaks with significant economic losses in livestock. Incursions of SPP and LSD have occurred in Europe. Vaccination with live attenuated homologous and heterologous viruses are routinely implemented to control these diseases. Using the gold standard virus neutralization test, we studied the ability of homologous and heterologous sera to neutralize the SPPV and LSDV. We found that LSD and SPP sera effectively neutralize their homologous viruses, and GTP sera can neutralize SPPV. However, while LSD sera effectively neutralizes SPPV, SPP and GTP sera cannot neutralize the LSDV to the same extent. We discuss the implications of these observations in disease assay methodology and heterologous vaccine efficacy.
Collapse
Affiliation(s)
- Francisco J. Berguido
- Animal Production and Health Laboratory, Joint FAO/IAEA Centre, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Wagramer Strasse 5, P.O. Box 100, 1400 Vienna, Austria (C.E.L.)
- Institute of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Richard Thiga Kangethe
- Animal Production and Health Laboratory, Joint FAO/IAEA Centre, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Wagramer Strasse 5, P.O. Box 100, 1400 Vienna, Austria (C.E.L.)
| | - Wendy Shell
- Institute for Veterinary Disease Control, AGES—Austrian Agency for Health and Food Safety, Robert Koch-Gasse 17, 2340 Mödling, Austria
| | - Viskam Wijewardana
- Animal Production and Health Laboratory, Joint FAO/IAEA Centre, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Wagramer Strasse 5, P.O. Box 100, 1400 Vienna, Austria (C.E.L.)
| | - Reingard Grabherr
- Institute of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Giovanni Cattoli
- Animal Production and Health Laboratory, Joint FAO/IAEA Centre, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Wagramer Strasse 5, P.O. Box 100, 1400 Vienna, Austria (C.E.L.)
| | - Charles Euloge Lamien
- Animal Production and Health Laboratory, Joint FAO/IAEA Centre, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Wagramer Strasse 5, P.O. Box 100, 1400 Vienna, Austria (C.E.L.)
| |
Collapse
|
5
|
Bennett RP, Yoluç Y, Salter JD, Ripp A, Jessen HJ, Kaiser SM, Smith HC. Sangivamycin is preferentially incorporated into viral RNA by the SARS-CoV-2 polymerase. Antiviral Res 2023; 218:105716. [PMID: 37690700 DOI: 10.1016/j.antiviral.2023.105716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/12/2023]
Abstract
Sangivamycin (S) is an adenosine (A) nucleoside analog with low nanomolar antiviral activity against SARS-CoV-2 in vitro. Previously, low nanomolar antiviral efficacy was revealed when tested against multiple viral variants in several cell types. SARS-CoV-2 RNA isolated from live virus infected cells and the virions released from these cells was analyzed by mass spectrometry (MS) for S incorporation. Dose-dependent incorporation occurred up to 1.8 S per 1,000 nucleotides (49 S per genome) throughout the viral genomes isolated from both infected cells and viral particles, but this incorporation did not change the viral mutation rate. In contrast, host mRNA, affinity purified from the same infected and treated cells, contained little or no S. Sangivamycin triphosphate (STP) was synthesized to evaluate its incorporation into RNA by recombinant SARS-CoV-2 RNA-dependent RNA polymerase (RdRp) under defined in vitro conditions. SARS-CoV-2 RdRp showed that S was not a chain terminator and S containing oligonucleotides templated as A. Though the antiviral mechanism remains to be determined, the data suggests that SARS-CoV-2 RdRp incorporates STP into SARS-CoV-2 RNA, which does not significantly impair viral RNA synthesis or the mutation rate.
Collapse
Affiliation(s)
| | - Yasemin Yoluç
- Department of Pharmaceutical Chemistry, Goethe University Frankfurt, Frankfurt, Germany.
| | | | - Alexander Ripp
- Institute of Organic Chemistry Albert-Ludwigs-University, Freiburg, Germany.
| | - Henning J Jessen
- Institute of Organic Chemistry Albert-Ludwigs-University, Freiburg, Germany.
| | - Stefanie M Kaiser
- Department of Pharmaceutical Chemistry, Goethe University Frankfurt, Frankfurt, Germany.
| | - Harold C Smith
- OyaGen, Inc., Rochester, NY, USA; Department of Biochemistry and Biophysics, Center for RNA Biology, Center for AIDS Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
6
|
Radion EI, Mukhin VE, Kholodova AV, Vladimirov IS, Alsaeva DY, Zhdanova AS, Ulasova NY, Bulanova NV, Makarov VV, Keskinov AA, Yudin SM. Functional Characteristics of Serum Anti-SARS-CoV-2 Antibodies against Delta and Omicron Variants after Vaccination with Sputnik V. Viruses 2023; 15:1349. [PMID: 37376648 DOI: 10.3390/v15061349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Anti-SARS-CoV-2 vaccination leads to the production of neutralizing as well as non-neutralizing antibodies. In the current study, we investigated the temporal dynamics of both sides of immunity after vaccination with two doses of Sputnik V against SARS-CoV-2 variants Wuhan-Hu-1 SARS-CoV-2 G614-variant (D614G), B.1.617.2 (Delta), and BA.1 (Omicron). First, we constructed a SARS-CoV-2 pseudovirus assay to assess the neutralization activity of vaccine sera. We show that serum neutralization activity against BA.1 compared to D614G is decreased by 8.16-, 11.05-, and 11.16- fold in 1, 4, and 6 months after vaccination, respectively. Moreover, previous vaccination did not increase serum neutralization activity against BA.1 in recovered patients. Next, we used the ADMP assay to evaluate the Fc-mediated function of vaccine-induced serum antibodies. Our results show that the antibody-dependent phagocytosis triggered by S-proteins of the D614G, B.1.617.2 and BA.1 variants did not differ significantly in vaccinated individuals. Moreover, the ADMP efficacy was retained over up to 6 months in vaccine sera. Our results demonstrate differences in the temporal dynamics of neutralizing and non-neutralizing antibody functions after vaccination with Sputnik V.
Collapse
Affiliation(s)
- Elizaveta I Radion
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Schukinskaya 5, Building 1, Moscow 123182, Russia
| | - Vladimir E Mukhin
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Schukinskaya 5, Building 1, Moscow 123182, Russia
| | - Alyona V Kholodova
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Schukinskaya 5, Building 1, Moscow 123182, Russia
| | - Ivan S Vladimirov
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Schukinskaya 5, Building 1, Moscow 123182, Russia
| | - Darya Y Alsaeva
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Schukinskaya 5, Building 1, Moscow 123182, Russia
| | - Anastasia S Zhdanova
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Schukinskaya 5, Building 1, Moscow 123182, Russia
| | - Natalya Y Ulasova
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Schukinskaya 5, Building 1, Moscow 123182, Russia
| | - Natalya V Bulanova
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Schukinskaya 5, Building 1, Moscow 123182, Russia
| | - Valentin V Makarov
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Schukinskaya 5, Building 1, Moscow 123182, Russia
| | - Anton A Keskinov
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Schukinskaya 5, Building 1, Moscow 123182, Russia
| | - Sergey M Yudin
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Schukinskaya 5, Building 1, Moscow 123182, Russia
| |
Collapse
|
7
|
Devaux CA, Fantini J. Unravelling Antigenic Cross-Reactions toward the World of Coronaviruses: Extent of the Stability of Shared Epitopes and SARS-CoV-2 Anti-Spike Cross-Neutralizing Antibodies. Pathogens 2023; 12:713. [PMID: 37242383 PMCID: PMC10220573 DOI: 10.3390/pathogens12050713] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
The human immune repertoire retains the molecular memory of a very great diversity of target antigens (epitopes) and can recall this upon a second encounter with epitopes against which it has previously been primed. Although genetically diverse, proteins of coronaviruses exhibit sufficient conservation to lead to antigenic cross-reactions. In this review, our goal is to question whether pre-existing immunity against seasonal human coronaviruses (HCoVs) or exposure to animal CoVs has influenced the susceptibility of human populations to SARS-CoV-2 and/or had an impact upon the physiopathological outcome of COVID-19. With the hindsight that we now have regarding COVID-19, we conclude that although antigenic cross-reactions between different coronaviruses exist, cross-reactive antibody levels (titers) do not necessarily reflect on memory B cell frequencies and are not always directed against epitopes which confer cross-protection against SARS-CoV-2. Moreover, the immunological memory of these infections is short-term and occurs in only a small percentage of the population. Thus, in contrast to what might be observed in terms of cross-protection at the level of a single individual recently exposed to circulating coronaviruses, a pre-existing immunity against HCoVs or other CoVs can only have a very minor impact on SARS-CoV-2 circulation at the level of human populations.
Collapse
Affiliation(s)
- Christian A. Devaux
- Laboratory Microbes Evolution Phylogeny and Infection (MEPHI), Aix-Marseille Université, IRD, APHM Institut Hospitalo-Universitaire—Méditerranée Infection, 13005 Marseille, France
- Centre National de la Recherche Scientifique (CNRS-SNC5039), 13009 Marseille, France
| | - Jacques Fantini
- Aix-Marseille Université, INSERM UMR_S 1072, 13015 Marseille, France
| |
Collapse
|
8
|
Fudolig M. Effect of Transmission and Vaccination on Time to Dominance of Emerging Viral Strains: A Simulation-Based Study. Microorganisms 2023; 11:microorganisms11040860. [PMID: 37110282 PMCID: PMC10144238 DOI: 10.3390/microorganisms11040860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
We studied the effect of transmissibility and vaccination on the time required for an emerging strain of an existing virus to dominate in the infected population using a simulation-based experiment. The emergent strain is assumed to be completely resistant to the available vaccine. A stochastic version of a modified SIR model for emerging viral strains was developed to simulate surveillance data for infections. The proportion of emergent viral strain infections among the infected was modeled using a logistic curve and the time to dominance (TTD) was recorded for each simulation. A factorial experiment was implemented to compare the TTD values for different transmissibility coefficients, vaccination rates, and initial vaccination coverage. We discovered a non-linear relationship between TTD and the relative transmissibility of the emergent strain for populations with low vaccination coverage. Furthermore, higher vaccination coverage and high vaccination rates in the population yielded significantly lower TTD values. Vaccinating susceptible individuals against the current strain increases the susceptible pool of the emergent virus, which leads to the emergent strain spreading faster and requiring less time to dominate the infected population.
Collapse
|
9
|
Frutos AM, Balmaseda A, Vydiswaran N, Patel M, Ojeda S, Brouwer A, Tutino R, Cai S, Bakker K, Sanchez N, Lopez R, Kuan G, Gordon A. Burden and seasonality of primary and secondary symptomatic common cold coronavirus infections in Nicaraguan children. Influenza Other Respir Viruses 2023; 17:e13078. [PMID: 36494188 PMCID: PMC9835451 DOI: 10.1111/irv.13086] [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: 11/15/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The current SARS-CoV-2 pandemic highlights the need for an increased understanding of coronavirus epidemiology. In a pediatric cohort in Nicaragua, we evaluate the seasonality and burden of common cold coronavirus (ccCoV) infection and evaluate likelihood of symptoms in reinfections. METHODS Children presenting with symptoms of respiratory illness were tested for each of the four ccCoVs (NL63, 229E, OC43, and HKU1). Annual blood samples collected before ccCoV infection were tested for antibodies against each ccCoV. Seasonality was evaluated using wavelet and generalized additive model (GAM) analyses, and age-period effects were investigated using a Poisson model. We also evaluate the risk of symptom presentation between primary and secondary infections. RESULTS In our cohort of 2576 children from 2011 to 2016, we observed 595 ccCoV infections and 107 cases of ccCoV-associated lower respiratory infection (LRI). The overall incidence rate was 61.1 per 1000 person years (95% confidence interval (CI): 56.3, 66.2). Children under two had the highest incidence of ccCoV infections and associated LRI. ccCoV incidence rapidly decreases until about age 6. Each ccCoV circulated throughout the year and demonstrated annual periodicity. Peaks of NL63 typically occurred 3 months before 229E peaks and 6 months after OC43 peaks. Approximately 69% of symptomatic ccCoV infections were secondary infections. There was slightly lower risk (rate ratio (RR): 0.90, 95% CI: 0.83, 0.97) of LRI between secondary and primary ccCoV infections among participants under the age of 5. CONCLUSIONS ccCoV spreads annually among children with the greatest burden among ages 0-1. Reinfection is common; prior infection is associated with slight protection against LRI among the youngest children.
Collapse
Affiliation(s)
- Aaron M. Frutos
- Department of Epidemiology, School of Public HealthUniversity of MichiganAnn ArborMichiganUSA
| | - Angel Balmaseda
- Health Center Sócrates Flores VivasMinistry of HealthManaguaNicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnósticoy ReferenciaMinistry of HealthManaguaNicaragua
| | - Nivea Vydiswaran
- Department of Epidemiology, School of Public HealthUniversity of MichiganAnn ArborMichiganUSA
| | - Mayuri Patel
- Department of Epidemiology, School of Public HealthUniversity of MichiganAnn ArborMichiganUSA
| | | | - Andrew Brouwer
- Department of Epidemiology, School of Public HealthUniversity of MichiganAnn ArborMichiganUSA
| | - Rebecca Tutino
- Department of Epidemiology, School of Public HealthUniversity of MichiganAnn ArborMichiganUSA
| | - Shuwei Cai
- Department of Epidemiology, School of Public HealthUniversity of MichiganAnn ArborMichiganUSA
| | - Kevin Bakker
- Department of Epidemiology, School of Public HealthUniversity of MichiganAnn ArborMichiganUSA
| | | | - Roger Lopez
- Laboratorio Nacional de Virología, Centro Nacional de Diagnósticoy ReferenciaMinistry of HealthManaguaNicaragua
- Sustainable Sciences InstituteManaguaNicaragua
| | - Guillermina Kuan
- Health Center Sócrates Flores VivasMinistry of HealthManaguaNicaragua
- Sustainable Sciences InstituteManaguaNicaragua
| | - Aubree Gordon
- Department of Epidemiology, School of Public HealthUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
10
|
Odendahl M, Endler I, Haubold B, Rodionov RN, Bornstein SR, Tonn T. SARS-CoV-2-specicific humoral immunity in convalescent patients with mild COVID-19 is supported by CD4 + T-cell help and negatively correlated with Alphacoronavirus-specific antibody titer. Immunol Lett 2022; 251-252:38-46. [PMID: 36174771 PMCID: PMC9512529 DOI: 10.1016/j.imlet.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/29/2022] [Accepted: 09/26/2022] [Indexed: 01/31/2023]
Abstract
This study aimed at investigating the nature of SARS-CoV-2-specific immunity in patients with mild COVID-19 and sought to identify parameters most relevant for the generation of neutralizing antibody responses in convalescent COVID-19 patients. In the majority of the examined patients a cellular as well as humoral immune response directed to SARS-CoV-2 was detected. The finding of an anti-SARS-CoV-2-reactive cellular immune response in healthy individuals suggests a pre-existing immunity to various common cold HCoVs which share close homology with SARS-CoV-2. The humoral immunity to the S protein of SARS-CoV-2 detected in convalescent COVID-19 patients correlates with the presence of SARS-CoV-2-reactive CD4+ T cells expressing Th1 cytokines. Remarkably, an inverse correlation of SARS-CoV-2 S protein-specific IgGs with HCoV-NL63 and HCoV-229E S1 protein-specific IgGs suggests that pre-existing immunity to Alphacoronaviruses might have had an inhibitory imprint on the immune response to SARS-CoV-2-infection in the examined patients with mild COVID-19.
Collapse
Affiliation(s)
- Marcus Odendahl
- Experimental Transfusion Medicine, Medical Faculty Carl Gustav Carus, Technical University Dresden, Germany,Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany,Corresponding author at: Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East gGmbH, Blasewitzerstr. 68-70, 01309 Dresden, Germany
| | - Iris Endler
- Experimental Transfusion Medicine, Medical Faculty Carl Gustav Carus, Technical University Dresden, Germany,Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Beate Haubold
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Roman N. Rodionov
- Department of Medicine III, University Hospital Carl-Gustav, Dresden, Germany
| | - Stefan R. Bornstein
- Department of Medicine III, University Hospital Carl-Gustav, Dresden, Germany,Department of Diabetes, School of Life Course Science and Medicine, King's College London, London, United Kingdom
| | - Torsten Tonn
- Experimental Transfusion Medicine, Medical Faculty Carl Gustav Carus, Technical University Dresden, Germany,Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany,Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| |
Collapse
|
11
|
Liu R, Lin X, Chen B, Hou Z, Zhang Q, Lin S, Geng L, Sun Z, Cao C, Shi Y, Xia X. The mutation features and geographical distributions of the surface glycoprotein (S gene) in SARS-CoV-2 strains: A comparative analysis of the early and current strains. J Med Virol 2022; 94:5363-5374. [PMID: 35871556 PMCID: PMC9350160 DOI: 10.1002/jmv.28023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/26/2022] [Accepted: 07/22/2022] [Indexed: 02/05/2023]
Abstract
The surface glycoprotein (S protein) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was used to develop coronavirus disease 2019 (COVID-19) vaccines. However, SARS-CoV-2, especially the S protein, has undergone rapid evolution and mutation, which has remained to be determined. Here, we analyzed and compared the early (12 237) and the current (more than 10 million) SARS-CoV-2 strains to identify the mutation features and geographical distribution of the S gene and S protein. Results showed that in the early strains, most of the loci were with relative low mutation frequency except S: 23403 (4486 strains), while in the current strains, there was a surge in the mutation strains and frequency, with S: 23403 constantly being the highest one, but tremendously increased to approximately 1050 times. Furthermore, D614 (S: 23403) was one of the most highly frequent mutations in the S protein of Omicron as of March 2022, and most of the mutant strains were still from the United States, and the United Kingdom. Further analysis demonstrated that in the receptor-binding domain, most of the loci with low mutation frequency in the early strains, while S: 22995 was nowadays the most prevalent loci with 3 122 491 strains in the current strains. Overall, we compare the mutation features of the S region in SARS-CoV-2 strains between the early and the current stains, providing insight into further studies in concert with emerging SARS-CoV-2 variants for COVID-19 vaccines.
Collapse
Affiliation(s)
- Rang Liu
- Center for Reproductive MedicinePeking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenGuangdongChina
- Shantou University Medical CollegeShantouGuangdongChina
| | - Xinran Lin
- Center for Reproductive MedicinePeking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenGuangdongChina
| | - Bing Chen
- Center for Reproductive MedicinePeking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenGuangdongChina
| | - Zhenhui Hou
- Center for Reproductive MedicinePeking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenGuangdongChina
| | - Qiuju Zhang
- Center for Reproductive MedicinePeking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenGuangdongChina
| | - Shouren Lin
- Center for Reproductive MedicinePeking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenGuangdongChina
| | - Lan Geng
- Center for Reproductive MedicinePeking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenGuangdongChina
| | - Zhongyi Sun
- Center for Reproductive MedicinePeking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenGuangdongChina
| | - Canhui Cao
- Center for Reproductive MedicinePeking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenGuangdongChina
- Department of Neurosurgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Yu Shi
- Center for Reproductive MedicinePeking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenGuangdongChina
| | - Xi Xia
- Center for Reproductive MedicinePeking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenGuangdongChina
| |
Collapse
|
12
|
Bold D, Roman-Sosa G, Gaudreault NN, Zayat B, Pogranichniy RM, Richt JA. Development of an Indirect ELISA for the Detection of SARS-CoV-2 Antibodies in Cats. Front Vet Sci 2022; 9:864884. [PMID: 35754530 PMCID: PMC9226769 DOI: 10.3389/fvets.2022.864884] [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: 01/28/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Companion animals are susceptible to a variety of coronaviruses, and recent studies show that felines are highly susceptible to SARS-CoV-2 infection. RT-PCR diagnostic is currently the method of choice to detect the presence of SARS-CoV-2-specific viral nucleic acids in animal samples during an active infection; however, serological assays are critical to determine whether animals were exposed to the virus and to determine the seroprevalence of SARS-CoV-2-specific antibodies in a defined population. In this study, we utilized recombinant nucleocapsid (N) protein and the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 expressed in E. coli (N) and mammalian cells (N, RBD) to develop indirect ELISA (iELISA) tests using well-characterized SARS-CoV-2-positive and -negative cat serum panels from previous experimental cat challenge studies. The optimal conditions for the iELISA tests were established based on checkerboard dilutions of antigens and antibodies. The diagnostic sensitivity for the detection of feline antibodies specific for the N or RBD proteins of the iELISA tests was between 93.3 and 97.8%, respectively, and the diagnostic specificity 95.5%. The iELISAs developed here can be used for high-throughput screening of cat sera for both antigens. The presence of SARS-CoV-2-specific antibodies in a BSL-2 biocontainment environment, unlike virus neutralization tests with live virus which have to be performed in BSL-3 laboratories.
Collapse
Affiliation(s)
- Dashzeveg Bold
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Gleyder Roman-Sosa
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
- Institut für Virologie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Giessen, Giessen, Germany
| | - Natasha N. Gaudreault
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Batsukh Zayat
- Institute of Veterinary Medicine, Mongolian University of Life Sciences, Ulaanbaatar, Mongolia
| | - Roman M. Pogranichniy
- Veterinary Diagnostic Laboratory, Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Juergen A. Richt
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
13
|
Han X, Ye Q. The variants of SARS-CoV-2 and the challenges of vaccines. J Med Virol 2022; 94:1366-1372. [PMID: 34890492 PMCID: PMC9015306 DOI: 10.1002/jmv.27513] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/13/2021] [Accepted: 12/07/2021] [Indexed: 12/15/2022]
Abstract
Since the outbreak of coronavirus disease 2019 (COVID-19), countries all over the world have suffered severe losses. It affects not only human life and health but also the economy. In response to COVID-19, countries have made tremendous efforts to vaccine development. The newly discovered variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have brought major challenges to the effectiveness and research of vaccines. This article reviews the existing literature and summarizes the main variants of the SARS-CoV-2 and its impact on vaccines, and provides new ideas for the later development of vaccines. An excellent job in developing and applying vaccines will be an important measure for epidemic prevention and control.
Collapse
Affiliation(s)
- Xiucui Han
- Department of Clinical Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthNational Children's Regional Medical CenterHangzhouZhejiangChina
| | - Qing Ye
- Department of Clinical Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthNational Children's Regional Medical CenterHangzhouZhejiangChina
| |
Collapse
|
14
|
Abstract
Adaptive immune responses play critical roles in viral clearance and protection against re-infection, and SARS-CoV-2 is no exception. What is exceptional is the rapid characterization of the immune response to the virus performed by researchers during the first 20 months of the pandemic. This has given us a more detailed understanding of SARS-CoV-2 compared to many viruses that have been with us for a long time. Furthermore, effective COVID-19 vaccines were developed in record time, and their rollout worldwide is already making a significant difference, although major challenges remain in terms of equal access. The pandemic has engaged scientists and the public alike, and terms such as seroprevalence, neutralizing antibodies, antibody escape and vaccine certificates have become familiar to a broad community. Here, we review key findings concerning B cell and antibody (Ab) responses to SARS-CoV-2, focusing on non-severe cases and anti-spike (S) Ab responses in particular, the latter being central to protective immunity induced by infection or vaccination. The emergence of viral variants that have acquired mutations in S acutely highlights the need for continued characterization of both emerging variants and Ab responses against these during the evolving pathogen-immune system arms race.
Collapse
Affiliation(s)
- Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Ols
- Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Karin Loré
- Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | | |
Collapse
|
15
|
Mutantu PN, Ngwe Tun MM, Nabeshima T, Yu F, Mukadi PK, Tanaka T, Tashiro M, Fujita A, Kanie N, Oshiro R, Takazono T, Imamura Y, Hirayama T, Moi ML, Inoue S, Izumikawa K, Yasuda J, Morita K. Development and Evaluation of Quantitative Immunoglobulin G Enzyme-Linked Immunosorbent Assay for the Diagnosis of Coronavirus Disease 2019 Using Truncated Recombinant Nucleocapsid Protein as Assay Antigen. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:9630. [PMID: 34574555 PMCID: PMC8469721 DOI: 10.3390/ijerph18189630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/04/2021] [Accepted: 09/09/2021] [Indexed: 11/24/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19). Real-time RT-PCR is the most commonly used method for COVID-19 diagnosis. However, serological assays are urgently needed as complementary tools to RT-PCR. Hachim et al. 2020 and Burbelo et al. 2020 demonstrated that anti-nucleocapsid(N) SARS-CoV-2 antibodies are higher and appear earlier than the spike antibodies. Additionally, cross-reactive antibodies against N protein are more prevalent than those against spike protein. We developed a less cross-reactive immunoglobulin G (IgG) indirect ELISA by using a truncated recombinant SARS-CoV-2 N protein as assay antigen. A highly conserved region of coronaviruses N protein was deleted and the protein was prepared using an E. coli protein expression system. A total of 177 samples collected from COVID-19 suspected cases and 155 negative control sera collected during the pre-COVID-19 period were applied to evaluate the assay's performance, with the plaque reduction neutralization test and the commercial SARS-CoV-2 spike protein IgG ELISA as gold standards. The SARS-CoV-2 N truncated protein-based ELISA showed similar sensitivity (91.1% vs. 91.9%) and specificity (93.8% vs. 93.8%) between the PRNT and spike IgG ELISA, as well as also higher specificity compared to the full-length N protein (93.8% vs. 89.9%). Our ELISA can be used for the diagnosis and surveillance of COVID-19.
Collapse
Affiliation(s)
- Pierre Nsele Mutantu
- Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (P.N.M.); (P.K.M.)
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (M.M.N.T.); (T.N.); (M.L.M.); (K.M.)
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Mya Myat Ngwe Tun
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (M.M.N.T.); (T.N.); (M.L.M.); (K.M.)
| | - Takeshi Nabeshima
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (M.M.N.T.); (T.N.); (M.L.M.); (K.M.)
| | - Fuxun Yu
- Guizhou Provincial People’s Hospital, Guiyang 550002, China;
| | - Patrick Kakoni Mukadi
- Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (P.N.M.); (P.K.M.)
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Takeshi Tanaka
- Infection Control and Education Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (M.T.); (A.F.); (K.I.)
| | - Masato Tashiro
- Infection Control and Education Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (M.T.); (A.F.); (K.I.)
| | - Ayumi Fujita
- Infection Control and Education Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (M.T.); (A.F.); (K.I.)
| | - Nobuhiro Kanie
- Department of Infectious Diseases, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (N.K.); (R.O.)
| | - Ryosaku Oshiro
- Department of Infectious Diseases, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (N.K.); (R.O.)
| | - Takahiro Takazono
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (Y.I.); (T.H.)
| | - Yoshifumi Imamura
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (Y.I.); (T.H.)
- Medical Education Development Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Tatsuro Hirayama
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (Y.I.); (T.H.)
| | - Meng Ling Moi
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (M.M.N.T.); (T.N.); (M.L.M.); (K.M.)
| | - Shingo Inoue
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (M.M.N.T.); (T.N.); (M.L.M.); (K.M.)
| | - Koichi Izumikawa
- Infection Control and Education Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (M.T.); (A.F.); (K.I.)
| | - Jiro Yasuda
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan;
| | - Kouichi Morita
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (M.M.N.T.); (T.N.); (M.L.M.); (K.M.)
| |
Collapse
|
16
|
Han X, Xu P, Ye Q. Analysis of COVID-19 vaccines: Types, thoughts, and application. J Clin Lab Anal 2021; 35:e23937. [PMID: 34396586 PMCID: PMC8418485 DOI: 10.1002/jcla.23937] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 07/06/2021] [Accepted: 07/22/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To deal with COVID-19, various countries have made many efforts, including the research and development of vaccines. The purpose of this manuscript was to summarize the development, application, and problems of COVID-19 vaccines. METHODS This article reviewed the existing literature to see the development of the COVID-19 vaccine. RESULTS We found that different types of vaccines had their own advantages and disadvantages. At the same time, the side effects of the vaccine, the dose of vaccination, the evaluation of the efficacy, and the application of the vaccine were all things worth studying. CONCLUSION The successful development of the COVID-19 vaccine concerns almost all countries and people in the world. We must do an excellent job of researching the immunogenicity and immune reactivity of the vaccines. We hope this review can help colleagues at home and abroad.
Collapse
Affiliation(s)
- Xiucui Han
- Department of Clinical LaboratoryThe Children’s HospitalZhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouChina
| | - Pengfei Xu
- Clinical LaboratoryZhejiang HospitalHangzhouChina
| | - Qing Ye
- Department of Clinical LaboratoryThe Children’s HospitalZhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouChina
| |
Collapse
|
17
|
Alnuqaydan AM, Almutary AG, Sukamaran A, Yang BTW, Lee XT, Lim WX, Ng YM, Ibrahim R, Darmarajan T, Nanjappan S, Chellian J, Candasamy M, Madheswaran T, Sharma A, Dureja H, Prasher P, Verma N, Kumar D, Palaniveloo K, Bisht D, Gupta G, Madan JR, Singh SK, Jha NK, Dua K, Chellappan DK. Middle East Respiratory Syndrome (MERS) Virus-Pathophysiological Axis and the Current Treatment Strategies. AAPS PharmSciTech 2021; 22:173. [PMID: 34105037 PMCID: PMC8186825 DOI: 10.1208/s12249-021-02062-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Middle East respiratory syndrome (MERS) is a lethal respiratory disease with its first case reported back in 2012 (Jeddah, Saudi Arabia). It is a novel, single-stranded, positive-sense RNA beta coronavirus (MERS-CoV) that was isolated from a patient who died from a severe respiratory illness. Later, it was found that this patient was infected with MERS. MERS is endemic to countries in the Middle East regions, such as Saudi Arabia, Jordan, Qatar, Oman, Kuwait and the United Arab Emirates. It has been reported that the MERS virus originated from bats and dromedary camels, the natural hosts of MERS-CoV. The transmission of the virus to humans has been thought to be either direct or indirect. Few camel-to-human transmissions were reported earlier. However, the mode of transmission of how the virus affects humans remains unanswered. Moreover, outbreaks in either family-based or hospital-based settings were observed with high mortality rates, especially in individuals who did not receive proper management or those with underlying comorbidities, such as diabetes and renal failure. Since then, there have been numerous reports hypothesising complications in fatal cases of MERS. Over the years, various diagnostic methods, treatment strategies and preventive measures have been strategised in containing the MERS infection. Evidence from multiple sources implicated that no treatment options and vaccines have been developed in specific, for the direct management of MERS-CoV infection. Nevertheless, there are supportive measures outlined in response to symptom-related management. Health authorities should stress more on infection and prevention control measures, to ensure that MERS remains as a low-level threat to public health.
Collapse
Affiliation(s)
- Abdullah M Alnuqaydan
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Abdulmajeed G Almutary
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Arulmalar Sukamaran
- School of Pharmacy, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Brian Tay Wei Yang
- School of Pharmacy, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Xiao Ting Lee
- School of Pharmacy, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Wei Xuan Lim
- School of Pharmacy, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Yee Min Ng
- School of Pharmacy, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Rania Ibrahim
- School of Health Sciences, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Thiviya Darmarajan
- School of Health Sciences, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Satheeshkumar Nanjappan
- Department of Natural Products, National Institute of Pharmaceutical Education & Research (NIPER-Kolkata), Chunilal Bhawan, Maniktala, Kolkata, West Bengal, 700054, India
| | - Jestin Chellian
- Department of Life Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Mayuren Candasamy
- Department of Life Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Thiagarajan Madheswaran
- Department of Pharmaceutical Technology, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Ankur Sharma
- Department of Life Science, School of Basic Science and Research, Sharda University, Knowledge Park, Uttar Pradesh, 201310, India
| | - Harish Dureja
- Faculty of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, 248007, India
| | - Nitin Verma
- Chitkara University School of Pharmacy, Chitkara University, Atal Shiksha Kunj, Atal Nagar, Himachal Pradesh, 174103, India
| | - Deepak Kumar
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Kishneth Palaniveloo
- Institute of Ocean and Earth Sciences, Institute for Advanced Studies Building, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Dheeraj Bisht
- Department of Pharmaceutical Sciences Bhimtal, Kumaun University Nainital, Uttarakhand, 263136, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, India
| | - Jyotsana R Madan
- Department of Pharmaceutics, Smt. Kashibai Navale College of Pharmacy, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
18
|
Vashishtha VM, Kumar P. Development of SARS-CoV-2 vaccines: challenges, risks, and the way forward. Hum Vaccin Immunother 2021; 17:1635-1649. [PMID: 33270478 PMCID: PMC7754925 DOI: 10.1080/21645515.2020.1845524] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/25/2020] [Accepted: 10/29/2020] [Indexed: 01/13/2023] Open
Abstract
The COVID-19 pandemic mandates the development of a safe and effective Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) vaccine. This review analyzes the complexities, challenges, and other vital issues associated with the development of the SARS-CoV-2 vaccine. A brief review of the immune responses (innate, antibody, and T-cell) to SARS-CoV-2, including immune targets, correlates of protection, and duration of immunity is presented. Approaches to vaccine development including different vaccine platforms, critical attributes of novel vaccine candidates, the status of the ongoing clinical trials, and the ways to speed up vaccine development are also reviewed. Despite a historical average success rate of only 6%, and a usual gestation period of 10-12 years for the development of a new vaccine, the world is on the verge of developing COVID-19 vaccines in an extraordinary short time span.
Collapse
Affiliation(s)
- Vipin M. Vashishtha
- Department of Pediatrics, Mangla Hospital & Research Center, Shakti Chowk, Bijnor, India
| | | |
Collapse
|
19
|
Hoffman T, Kolstad L, Lindahl JF, Albinsson B, Bergqvist A, Rönnberg B, Lundkvist Å. Diagnostic Potential of a Luminex-Based Coronavirus Disease 2019 Suspension Immunoassay (COVID-19 SIA) for the Detection of Antibodies against SARS-CoV-2. Viruses 2021; 13:v13060993. [PMID: 34073484 PMCID: PMC8227055 DOI: 10.3390/v13060993] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
Due to the current, rapidly increasing Coronavirus disease 2019 (COVID-19) pandemic, efficient and highly specific diagnostic methods are needed. The receptor-binding part of the spike (S) protein, S1, has been suggested to be highly virus-specific; it does not cross-react with antibodies against other coronaviruses. Three recombinant partial S proteins of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) expressed in mammalian or baculovirus-insect cells were evaluated as antigens in a Luminex-based suspension immunoassay (SIA). The best performing antigen (S1; amino acids 16-685) was selected and further evaluated by serum samples from 76 Swedish patients or convalescents with COVID-19 (previously PCR and/or serologically confirmed), 200 pre-COVID-19 individuals (180 blood donors and 20 infants), and 10 patients with acute Epstein-Barr virus infection. All 76 positive samples showed detectable antibodies to S1, while none of the 210 negative controls gave a false positive antibody reaction. We further compared the COVID-19 SIA with a commercially available enzyme immunoassay and a previously evaluated COVID-19 rapid antibody test. The results revealed an overall assay sensitivity of 100%, a specificity of 100% for both IgM and IgG, a quantitative ability at concentrations up to 25 BAU/mL, and a better performance as compared to the commercial assays, suggesting the COVID-19 SIA as a most valuable tool for efficient laboratory-based serology.
Collapse
Affiliation(s)
- Tove Hoffman
- Department of Medical Biochemistry and Microbiology, Zoonosis Science Centre, Uppsala University, Husargatan 3, SE-751 23 Uppsala, Sweden; (L.K.); (J.F.L.); (B.A.); (B.R.); (Å.L.)
- Correspondence:
| | - Linda Kolstad
- Department of Medical Biochemistry and Microbiology, Zoonosis Science Centre, Uppsala University, Husargatan 3, SE-751 23 Uppsala, Sweden; (L.K.); (J.F.L.); (B.A.); (B.R.); (Å.L.)
| | - Johanna F. Lindahl
- Department of Medical Biochemistry and Microbiology, Zoonosis Science Centre, Uppsala University, Husargatan 3, SE-751 23 Uppsala, Sweden; (L.K.); (J.F.L.); (B.A.); (B.R.); (Å.L.)
| | - Bo Albinsson
- Department of Medical Biochemistry and Microbiology, Zoonosis Science Centre, Uppsala University, Husargatan 3, SE-751 23 Uppsala, Sweden; (L.K.); (J.F.L.); (B.A.); (B.R.); (Å.L.)
- Laboratory of Clinical Microbiology, Uppsala University Hospital, Dag Hammarskjölds väg 38, SE-752 37 Uppsala, Sweden;
| | - Anders Bergqvist
- Laboratory of Clinical Microbiology, Uppsala University Hospital, Dag Hammarskjölds väg 38, SE-752 37 Uppsala, Sweden;
- Department of Medical Sciences, Uppsala University, Dag Hammarskjölds väg 38, SE-751 84 Uppsala, Sweden
| | - Bengt Rönnberg
- Department of Medical Biochemistry and Microbiology, Zoonosis Science Centre, Uppsala University, Husargatan 3, SE-751 23 Uppsala, Sweden; (L.K.); (J.F.L.); (B.A.); (B.R.); (Å.L.)
- Laboratory of Clinical Microbiology, Uppsala University Hospital, Dag Hammarskjölds väg 38, SE-752 37 Uppsala, Sweden;
| | - Åke Lundkvist
- Department of Medical Biochemistry and Microbiology, Zoonosis Science Centre, Uppsala University, Husargatan 3, SE-751 23 Uppsala, Sweden; (L.K.); (J.F.L.); (B.A.); (B.R.); (Å.L.)
| |
Collapse
|
20
|
Pietschmann J, Voepel N, Voß L, Rasche S, Schubert M, Kleines M, Krause HJ, Shaw TM, Spiegel H, Schroeper F. Development of Fast and Portable Frequency Magnetic Mixing-Based Serological SARS-CoV-2-Specific Antibody Detection Assay. Front Microbiol 2021; 12:643275. [PMID: 34025604 PMCID: PMC8132704 DOI: 10.3389/fmicb.2021.643275] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/23/2021] [Indexed: 01/11/2023] Open
Abstract
A novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) emerged in China in December 2019, causing an ongoing, rapidly spreading global pandemic. Worldwide, vaccination is now expected to provide containment of the novel virus, resulting in an antibody-mediated immunity. To verify this, serological antibody assays qualitatively as well as quantitatively depicting the amount of generated antibodies are of great importance. Currently available test methods are either laboratory based or do not have the ability to indicate an estimation about the immune response. To overcome this, a novel and rapid serological magnetic immunodetection (MID) point-of-care (PoC) assay was developed, with sensitivity and specificity comparable to laboratory-based DiaSorin Liaison SARS-CoV-2 S1/S2 IgG assay. To specifically enrich human antibodies against SARS-CoV-2 in immunofiltration columns (IFCs) from patient sera, a SARS-CoV-2 S1 antigen was transiently produced in plants, purified and immobilized on the IFC. Then, an IgG-specific secondary antibody could bind to the retained antibodies, which was finally labeled using superparamagnetic nanoparticles. Based on frequency magnetic mixing technology (FMMD), the magnetic particles enriched in IFC were detected using a portable FMMD device. The obtained measurement signal correlates with the amount of SARS-CoV-2-specific antibodies in the sera, which could be demonstrated by titer determination. In this study, a MID-based assay could be developed, giving qualitative as well as semiquantitative results of SARS-CoV-2-specific antibody levels in patient's sera within 21 min of assay time with a sensitivity of 97% and a specificity of 92%, based on the analysis of 170 sera from hospitalized patients that were tested using an Food and Drug Administration (FDA)-certified chemiluminescence assay.
Collapse
Affiliation(s)
- Jan Pietschmann
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Nadja Voepel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Leonie Voß
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Stefan Rasche
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Max Schubert
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Michael Kleines
- Laboratory Diagnostic Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Hans-Joachim Krause
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich, Jülich, Germany
| | | | - Holger Spiegel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Florian Schroeper
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| |
Collapse
|
21
|
Gunn BM, Lu R, Slein MD, Ilinykh PA, Huang K, Atyeo C, Schendel SL, Kim J, Cain C, Roy V, Suscovich TJ, Takada A, Halfmann PJ, Kawaoka Y, Pauthner MG, Momoh M, Goba A, Kanneh L, Andersen KG, Schieffelin JS, Grant D, Garry RF, Saphire EO, Bukreyev A, Alter G. A Fc engineering approach to define functional humoral correlates of immunity against Ebola virus. Immunity 2021; 54:815-828.e5. [PMID: 33852832 PMCID: PMC8111768 DOI: 10.1016/j.immuni.2021.03.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 01/28/2021] [Accepted: 03/16/2021] [Indexed: 01/31/2023]
Abstract
Protective Ebola virus (EBOV) antibodies have neutralizing activity and induction of antibody constant domain (Fc)-mediated innate immune effector functions. Efforts to enhance Fc effector functionality often focus on maximizing antibody-dependent cellular cytotoxicity, yet distinct combinations of functions could be critical for antibody-mediated protection. As neutralizing antibodies have been cloned from EBOV disease survivors, we sought to identify survivor Fc effector profiles to help guide Fc optimization strategies. Survivors developed a range of functional antibody responses, and we therefore applied a rapid, high-throughput Fc engineering platform to define the most protective profiles. We generated a library of Fc variants with identical antigen-binding fragments (Fabs) from an EBOV neutralizing antibody. Fc variants with antibody-mediated complement deposition and moderate natural killer (NK) cell activity demonstrated complete protective activity in a stringent in vivo mouse model. Our findings highlight the importance of specific effector functions in antibody-mediated protection, and the experimental platform presents a generalizable resource for identifying correlates of immunity to guide therapeutic antibody design.
Collapse
Affiliation(s)
- Bronwyn M Gunn
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Richard Lu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Matthew D Slein
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Philipp A Ilinykh
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Kai Huang
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Caroline Atyeo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | - Jiyoung Kim
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Caitlin Cain
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Vicky Roy
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | - Ayato Takada
- Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Peter J Halfmann
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Matthias G Pauthner
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| | - Mambu Momoh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone
| | - Augustine Goba
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone
| | - Lansana Kanneh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone
| | - Kristian G Andersen
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA; Scripps Research Translational Institute, La Jolla, CA, USA
| | - John S Schieffelin
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, USA
| | - Donald Grant
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone; Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Robert F Garry
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA; Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, USA.
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
22
|
Benzigar MR, Bhattacharjee R, Baharfar M, Liu G. Current methods for diagnosis of human coronaviruses: pros and cons. Anal Bioanal Chem 2021; 413:2311-2330. [PMID: 33219449 PMCID: PMC7679240 DOI: 10.1007/s00216-020-03046-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/14/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022]
Abstract
The current global fight against coronavirus disease (COVID-19) to flatten the transmission curve is put forth by the World Health Organization (WHO) as there is no immediate diagnosis or cure for COVID-19 so far. In order to stop the spread, researchers worldwide are working around the clock aiming to develop reliable tools for early diagnosis of severe acute respiratory syndrome (SARS-CoV-2) understanding the infection path and mechanisms. Currently, nucleic acid-based molecular diagnosis (real-time reverse transcription polymerase chain reaction (RT-PCR) test) is considered the gold standard for early diagnosis of SARS-CoV-2. Antibody-based serology detection is ineffective for the purpose of early diagnosis, but a potential tool for serosurveys, providing people with immune certificates for clearance from COVID-19 infection. Meanwhile, there are various blooming methods developed these days. In this review, we summarise different types of coronavirus discovered which can be transmitted between human beings. Methods used for diagnosis of the discovered human coronavirus (SARS, MERS, COVID-19) including nucleic acid detection, gene sequencing, antibody detection, antigen detection, and clinical diagnosis are presented. Their merits, demerits and prospects are discussed which can help the researchers to develop new generation of advanced diagnostic tools for accurate and effective control of human coronavirus transmission in the communities and hospitals.
Collapse
Affiliation(s)
- Mercy R Benzigar
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Ripon Bhattacharjee
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Mahroo Baharfar
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Guozhen Liu
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia.
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
23
|
Ansari A, Arya R, Sachan S, Jha SN, Kalia A, Lall A, Sette A, Grifoni A, Weiskopf D, Coshic P, Sharma A, Gupta N. Immune Memory in Mild COVID-19 Patients and Unexposed Donors Reveals Persistent T Cell Responses After SARS-CoV-2 Infection. Front Immunol 2021; 12:636768. [PMID: 33777028 PMCID: PMC7991090 DOI: 10.3389/fimmu.2021.636768] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/25/2021] [Indexed: 12/24/2022] Open
Abstract
Understanding the causes of the diverse outcome of COVID-19 pandemic in different geographical locations is important for the worldwide vaccine implementation and pandemic control responses. We analyzed 42 unexposed healthy donors and 28 mild COVID-19 subjects up to 5 months from the recovery for SARS-CoV-2 specific immunological memory. Using HLA class II predicted peptide megapools, we identified SARS-CoV-2 cross-reactive CD4+ T cells in around 66% of the unexposed individuals. Moreover, we found detectable immune memory in mild COVID-19 patients several months after recovery in the crucial arms of protective adaptive immunity; CD4+ T cells and B cells, with a minimal contribution from CD8+ T cells. Interestingly, the persistent immune memory in COVID-19 patients is predominantly targeted towards the Spike glycoprotein of the SARS-CoV-2. This study provides the evidence of both high magnitude pre-existing and persistent immune memory in Indian population. By providing the knowledge on cellular immune responses to SARS-CoV-2, our work has implication for the development and implementation of vaccines against COVID-19.
Collapse
Affiliation(s)
- Asgar Ansari
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, India
| | - Rakesh Arya
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Shilpa Sachan
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, India
| | - Someshwar Nath Jha
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, India
| | - Anurag Kalia
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, India
| | - Anupam Lall
- Department of Transfusion Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States.,Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, United States
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Poonam Coshic
- Department of Transfusion Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Nimesh Gupta
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
24
|
Ansari A, Arya R, Sachan S, Jha SN, Kalia A, Lall A, Sette A, Grifoni A, Weiskopf D, Coshic P, Sharma A, Gupta N. Immune memory in mild COVID-19 patients and unexposed donors from India reveals persistent T cell responses after SARS-CoV-2 infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2020.11.16.20232967. [PMID: 33655267 PMCID: PMC7924292 DOI: 10.1101/2020.11.16.20232967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Understanding the causes of the diverse outcome of COVID-19 pandemic in different geographical locations is important for the worldwide vaccine implementation and pandemic control responses. We analyzed 42 unexposed healthy donors and 28 mild COVID-19 subjects up to 5 months from the recovery for SARS-CoV-2 specific immunological memory. Using HLA class II predicted peptide megapools, we identified SARS-CoV-2 cross-reactive CD4+ T cells in around 66% of the unexposed individuals. Moreover, we found detectable immune memory in mild COVID-19 patients several months after recovery in the crucial arms of protective adaptive immunity; CD4+ T cells and B cells, with a minimal contribution from CD8+ T cells. Interestingly, the persistent immune memory in COVID-19 patients is predominantly targeted towards the Spike glycoprotein of the SARS-CoV-2. This study provides the evidence of both high magnitude pre-existing and persistent immune memory in Indian population. By providing the knowledge on cellular immune responses to SARS-CoV-2, our work has implication for the development and implementation of vaccines against COVID-19.
Collapse
Affiliation(s)
- Asgar Ansari
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Rakesh Arya
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Shilpa Sachan
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Someshwar Nath Jha
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Anurag Kalia
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Anupam Lall
- Department of Transfusion Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Poonam Coshic
- Department of Transfusion Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Nimesh Gupta
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, 110067, India
| |
Collapse
|
25
|
Sherwood LJ, Hayhurst A. Toolkit for Quickly Generating and Characterizing Molecular Probes Specific for SARS-CoV-2 Nucleocapsid as a Primer for Future Coronavirus Pandemic Preparedness. ACS Synth Biol 2021; 10:379-390. [PMID: 33534552 PMCID: PMC7875338 DOI: 10.1021/acssynbio.0c00566] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Indexed: 12/31/2022]
Abstract
Generating and characterizing immunoreagents to enable studies of novel emerging viruses is an area where ensembles of synthetic genes, recombinant antibody pipelines, and modular antibody-reporter fusion proteins can respond rapidly. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread through the global population causing widespread morbidity, mortality, and socioeconomic chaos. Using SARS-CoV-2 as our model and starting with a gBlocks encoded nucleocapsid (N) gene, we purified recombinant protein from E. coli, to serve as bait for selecting semisynthetic nanobodies from our Nomad single-pot library. Clones were isolated in days and first fused to Gaussia luciferase to determine EC50 in the tens of nM range, and second fused to the ascorbate peroxidase derivative APEX2 for sensitive detection of SARS-CoV-2 infected cells. To generate inherently fluorescent immunoreagents, we introduce novel periplasmic sdAb fusions made with mNeonGreen and mScarlet-I, which were produced at milligram amounts. The fluorescent fusion proteins enabled concise visualization of SARS-CoV-2 N in the cytoplasm but not in the nucleus 24 h post infection, akin to the distribution of SARS-CoV N, thereby validating these useful imaging tools. SdAb reactivity appeared specific to SARS-CoV-2 with very much weaker binding to SARS-CoV, and no noticeable cross-reactivity to a panel of overexpressed human codon optimized N proteins from other CoV. High periplasmic expression levels and in silico immortalization of the nanobody constructs guarantees a cost-effective and reliable source of SARS-CoV-2 immunoreagents. Our proof-of-principle study should be applicable to known and newly emerging CoV to broaden the tools available for their analysis and help safeguard human health in a more proactive than reactive manner.
Collapse
Affiliation(s)
- Laura Jo Sherwood
- Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Andrew Hayhurst
- Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| |
Collapse
|
26
|
de Assis RR, Jain A, Nakajima R, Jasinskas A, Felgner J, Obiero JM, Norris PJ, Stone M, Simmons G, Bagri A, Irsch J, Schreiber M, Buser A, Holbro A, Battegay M, Hosimer P, Noesen C, Adenaiye O, Tai S, Hong F, Milton DK, Davies DH, Contestable P, Corash LM, Busch MP, Felgner PL, Khan S. Analysis of SARS-CoV-2 antibodies in COVID-19 convalescent blood using a coronavirus antigen microarray. Nat Commun 2021; 12:6. [PMID: 33397903 PMCID: PMC7782488 DOI: 10.1038/s41467-020-20095-2] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 11/10/2020] [Indexed: 01/28/2023] Open
Abstract
The current practice for diagnosis of COVID-19, based on SARS-CoV-2 PCR testing of pharyngeal or respiratory specimens in a symptomatic patient at high epidemiologic risk, likely underestimates the true prevalence of infection. Serologic methods can more accurately estimate the disease burden by detecting infections missed by the limited testing performed to date. Here, we describe the validation of a coronavirus antigen microarray containing immunologically significant antigens from SARS-CoV-2, in addition to SARS-CoV, MERS-CoV, common human coronavirus strains, and other common respiratory viruses. A comparison of antibody profiles detected on the array from control sera collected prior to the SARS-CoV-2 pandemic versus convalescent blood specimens from virologically confirmed COVID-19 cases demonstrates near complete discrimination of these two groups, with improved performance from use of antigen combinations that include both spike protein and nucleoprotein. This array can be used as a diagnostic tool, as an epidemiologic tool to more accurately estimate the disease burden of COVID-19, and as a research tool to correlate antibody responses with clinical outcomes.
Collapse
Affiliation(s)
- Rafael R de Assis
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Aarti Jain
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Rie Nakajima
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Algis Jasinskas
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Jiin Felgner
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Joshua M Obiero
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Philip J Norris
- Vitalant Research Institute, San Francisco, CA, USA.,Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Mars Stone
- Vitalant Research Institute, San Francisco, CA, USA.,Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Graham Simmons
- Vitalant Research Institute, San Francisco, CA, USA.,Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | | | | | - Martin Schreiber
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Andreas Buser
- Division of Infectious Diseases & Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Andreas Holbro
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Manuel Battegay
- Division of Infectious Diseases & Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | | | | | - Oluwasanmi Adenaiye
- Institute for Applied Environmental Health, School of Public Health, University of Maryland, College Park, MD, USA
| | - Sheldon Tai
- Institute for Applied Environmental Health, School of Public Health, University of Maryland, College Park, MD, USA
| | - Filbert Hong
- Institute for Applied Environmental Health, School of Public Health, University of Maryland, College Park, MD, USA
| | - Donald K Milton
- Institute for Applied Environmental Health, School of Public Health, University of Maryland, College Park, MD, USA
| | - D Huw Davies
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | | | | | - Michael P Busch
- Vitalant Research Institute, San Francisco, CA, USA.,Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Philip L Felgner
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Saahir Khan
- Division of Infectious Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
27
|
Huang T, Sun L, Kang D, Poongavanam V, Liu X, Zhan P, Menéndez-Arias L. Search, Identification, and Design of Effective Antiviral Drugs Against Pandemic Human Coronaviruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1322:219-260. [PMID: 34258743 DOI: 10.1007/978-981-16-0267-2_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Recent coronavirus outbreaks of SARS-CoV-1 (2002-2003), MERS-CoV (since 2012), and SARS-CoV-2 (since the end of 2019) are examples of how viruses can damage health care and generate havoc all over the world. Coronavirus can spread quickly from person to person causing high morbidity and mortality. Unfortunately, the antiviral armamentarium is insufficient to fight these infections. In this chapter, we provide a detailed summary of the current situation in the development of drugs directed against pandemic human coronaviruses. Apart from the recently licensed remdesivir, other antiviral agents discussed in this review include molecules targeting viral components (e.g., RNA polymerase inhibitors, entry inhibitors, or protease inhibitors), compounds interfering with virus-host interactions, and drugs identified in large screening assays, effective against coronavirus replication, but with an uncertain mechanism of action.
Collapse
Affiliation(s)
- Tianguang Huang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Lin Sun
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | | | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Luis Menéndez-Arias
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
28
|
Zhao J, Zhao S, Ou J, Zhang J, Lan W, Guan W, Wu X, Yan Y, Zhao W, Wu J, Chodosh J, Zhang Q. COVID-19: Coronavirus Vaccine Development Updates. Front Immunol 2020; 11:602256. [PMID: 33424848 PMCID: PMC7785583 DOI: 10.3389/fimmu.2020.602256] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/26/2020] [Indexed: 12/27/2022] Open
Abstract
Coronavirus Disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), a newly emerged coronavirus, and has been pandemic since March 2020 and led to many fatalities. Vaccines represent the most efficient means to control and stop the pandemic of COVID-19. However, currently there is no effective COVID-19 vaccine approved to use worldwide except for two human adenovirus vector vaccines, three inactivated vaccines, and one peptide vaccine for early or limited use in China and Russia. Safe and effective vaccines against COVID-19 are in urgent need. Researchers around the world are developing 213 COVID-19 candidate vaccines, among which 44 are in human trials. In this review, we summarize and analyze vaccine progress against SARS-CoV, Middle-East respiratory syndrome Coronavirus (MERS-CoV), and SARS-CoV-2, including inactivated vaccines, live attenuated vaccines, subunit vaccines, virus like particles, nucleic acid vaccines, and viral vector vaccines. As SARS-CoV-2, SARS-CoV, and MERS-CoV share the common genus, Betacoronavirus, this review of the major research progress will provide a reference and new insights into the COVID-19 vaccine design and development.
Collapse
Affiliation(s)
- Jing Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Shan Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Junxian Ou
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jing Zhang
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Wendong Lan
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wenyi Guan
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaowei Wu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yuqian Yan
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wei Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jianguo Wu
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - James Chodosh
- Department of Ophthalmology, Howe Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Qiwei Zhang
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| |
Collapse
|
29
|
Li YD, Chi WY, Su JH, Ferrall L, Hung CF, Wu TC. Coronavirus vaccine development: from SARS and MERS to COVID-19. J Biomed Sci 2020; 27:104. [PMID: 33341119 PMCID: PMC7749790 DOI: 10.1186/s12929-020-00695-2] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/20/2020] [Indexed: 02/08/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a new type of coronavirus that causes the Coronavirus Disease 2019 (COVID-19), which has been the most challenging pandemic in this century. Considering its high mortality and rapid spread, an effective vaccine is urgently needed to control this pandemic. As a result, the academia, industry, and government sectors are working tightly together to develop and test a variety of vaccines at an unprecedented pace. In this review, we outline the essential coronavirus biological characteristics that are important for vaccine design. In addition, we summarize key takeaways from previous vaccination studies of Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and Middle East Respiratory Syndrome Coronavirus (MERS-CoV), highlighting the pros and cons of each immunization strategy. Finally, based on these prior vaccination experiences, we discuss recent progress and potential challenges of COVID-19 vaccine development.
Collapse
Affiliation(s)
- Yen-Der Li
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Wei-Yu Chi
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jun-Han Su
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Louise Ferrall
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Chien-Fu Hung
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - T-C Wu
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
- Johns Hopkins School of Medicine, 1550 Orleans St, CRB II - Room 309, Baltimore, MD, 21287, USA.
| |
Collapse
|
30
|
Harvala H, Robb ML, Watkins N, Ijaz S, Dicks S, Patel M, Supasa P, Wanwisa D, Liu C, Mongkolsapaya J, Bown A, Bailey D, Vipond R, Grayson N, Temperton N, Gupta S, Ploeg RJ, Bolton J, Fyfe A, Gopal R, Simmonds P, Screaton G, Thompson C, Brooks T, Zambon M, Miflin G, Roberts DJ. Convalescent plasma therapy for the treatment of patients with COVID-19: Assessment of methods available for antibody detection and their correlation with neutralising antibody levels. Transfus Med 2020; 31:167-175. [PMID: 33333627 PMCID: PMC8246874 DOI: 10.1111/tme.12746] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/26/2022]
Abstract
INTRODUCTION The lack of approved specific therapeutic agents to treat coronavirus disease (COVID-19) associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has led to the rapid implementation of convalescent plasma therapy (CPT) trials in many countries, including the United Kingdom. Effective CPT is likely to require high titres of neutralising antibody (nAb) in convalescent donations. Understanding the relationship between functional neutralising antibodies and antibody levels to specific SARS-CoV-2 proteins in scalable assays will be crucial for the success of a large-scale collection. We assessed whether neutralising antibody titres correlated with reactivity in a range of enzyme-linked immunosorbent assays (ELISA) targeting the spike (S) protein, the main target for human immune response. METHODS Blood samples were collected from 52 individuals with a previous laboratory-confirmed SARS-CoV-2 infection. These were assayed for SARS-CoV-2 nAbs by microneutralisation and pseudo-type assays and for antibodies by four different ELISAs. Receiver operating characteristic (ROC) analysis was used to further identify sensitivity and specificity of selected assays to identify samples containing high nAb levels. RESULTS All samples contained SARS-CoV-2 antibodies, whereas neutralising antibody titres of greater than 1:20 were detected in 43 samples (83% of those tested) and >1:100 in 22 samples (42%). The best correlations were observed with EUROimmun immunoglobulin G (IgG) reactivity (Spearman Rho correlation coefficient 0.88; p < 0.001). Based on ROC analysis, EUROimmun would detect 60% of samples with titres of >1:100 with 100% specificity using a reactivity index of 9.1 (13/22). DISCUSSION Robust associations between nAb titres and reactivity in several ELISA-based antibody tests demonstrate their possible utility for scaled-up production of convalescent plasma containing potentially therapeutic levels of anti-SARS-CoV-2 nAbs.
Collapse
Affiliation(s)
- Heli Harvala
- National Microbiology Services, NHS Blood and Transplant, London, UK
| | - Matthew L Robb
- Statistics and Clinical Studies, NHS Blood and Transplant, Bristol, UK
| | - Nick Watkins
- Department of Research and Development, NHS Blood and Transplant Cambridge, Cambridge, UK
| | - Samreen Ijaz
- Virology Reference Department, National Infection Service, Public Health England, London, UK
| | - Steven Dicks
- Virology Reference Department, National Infection Service, Public Health England, London, UK
| | - Monika Patel
- High Containment Microbiology, National Infection Service, Public Health England, London, UK
| | - Piyada Supasa
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Dejnirattisai Wanwisa
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Chang Liu
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Juthathip Mongkolsapaya
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Dengue Hemorrhagic Fever Research Unit, Office for Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Abbie Bown
- Rare and Imported Pathogens Laboratory, Public Health England, Porton Down, Wiltshire, UK
| | - Daniel Bailey
- Rare and Imported Pathogens Laboratory, Public Health England, Porton Down, Wiltshire, UK
| | - Richard Vipond
- Rare and Imported Pathogens Laboratory, Public Health England, Porton Down, Wiltshire, UK
| | - Nicholas Grayson
- Department of Paediatric Medicine, University of Oxford, University of Oxford, Oxford, UK
| | | | - Sunetra Gupta
- Department of Zoology, University of Oxford, Oxford, UK
| | - Rutger J Ploeg
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.,Department of Transplant Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jai Bolton
- Department of Zoology, University of Oxford, Oxford, UK
| | - Alex Fyfe
- Department of Zoology, University of Oxford, Oxford, UK
| | - Robin Gopal
- Virology Reference Department, National Infection Service, Public Health England, London, UK
| | - Peter Simmonds
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Gavin Screaton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Tim Brooks
- Rare and Imported Pathogens Laboratory, Public Health England, Porton Down, Wiltshire, UK
| | - Maria Zambon
- Virology Reference Department, National Infection Service, Public Health England, London, UK
| | - Gail Miflin
- Department of Chief Medical Officer, NHS Blood and Transplant, Bristol, UK
| | - David J Roberts
- NHS Blood and Transplant, Oxford, John Radcliffe Hospital, Oxford, UK.,Radcliffe Department of Medicine and BRC Haematology Theme, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
31
|
Canedo-Marroquín G, Saavedra F, Andrade CA, Berrios RV, Rodríguez-Guilarte L, Opazo MC, Riedel CA, Kalergis AM. SARS-CoV-2: Immune Response Elicited by Infection and Development of Vaccines and Treatments. Front Immunol 2020; 11:569760. [PMID: 33362758 PMCID: PMC7759609 DOI: 10.3389/fimmu.2020.569760] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/13/2020] [Indexed: 01/08/2023] Open
Abstract
The World Health Organization (WHO) announced in March a pandemic caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). This new infectious disease was named Coronavirus Disease 19 (COVID-19), and at October 2020, more than 39,000,000 cases of SARS-CoV-2 have been detected worldwide leading to near 1,100,000 deaths. Clinically, COVID-19 is characterized by clinical manifestations, such as fever, dry cough, headache, and in more severe cases, respiratory distress. Moreover, neurological-, cardiac-, and renal-related symptoms have also been described. Clinical evidence suggests that migration of immune cells to the affected organs can produce an exacerbated release of proinflammatory mediators that contribute to disease and render the immune response as a major player during the development of the COVID-19 disease. Due to the current sanitary situation, the development of vaccines is imperative. Up to the date, 42 prototypes are being tested in humans in different clinical stages, with 10 vaccine candidates undergoing evaluation in phase III clinical trials. In the same way, the search for an effective treatment to approach the most severe cases is also in constant advancement. Several potential therapies have been tested since COVID-19 was described, including antivirals, antiparasitic and immune modulators. Recently, clinical trials with hydroxychloroquine-a promising drug in the beginning-were suspended. In addition, the Food and Drug Administration (FDA) approved convalescent serum administration as a treatment for SARS-CoV-2 patients. Moreover, monoclonal antibody therapy is also under development to neutralize the virus and prevent infection. In this article, we describe the clinical manifestations and the immunological information available about COVID-19 disease. Furthermore, we discuss current therapies under study and the development of vaccines to prevent this disease.
Collapse
Affiliation(s)
- Gisela Canedo-Marroquín
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Farides Saavedra
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A. Andrade
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roslye V. Berrios
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Linmar Rodríguez-Guilarte
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María C. Opazo
- Millennium Institute on Immunology and Immunotherapy Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Claudia A. Riedel
- Millennium Institute on Immunology and Immunotherapy Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
32
|
Rostad CA, Chahroudi A, Mantus G, Lapp SA, Teherani M, Macoy L, Tarquinio KM, Basu RK, Kao C, Linam WM, Zimmerman MG, Shi PY, Menachery VD, Oster ME, Edupuganti S, Anderson EJ, Suthar MS, Wrammert J, Jaggi P. Quantitative SARS-CoV-2 Serology in Children With Multisystem Inflammatory Syndrome (MIS-C). Pediatrics 2020; 146:peds.2020-018242. [PMID: 32879033 DOI: 10.1542/peds.2020-018242] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/27/2020] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVES We aimed to measure severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) serological responses in children hospitalized with multisystem inflammatory syndrome in children (MIS-C) compared with those with coronavirus disease 2019 (COVID-19), those with Kawasaki disease (KD), and hospitalized pediatric controls. METHODS From March 17, 2020, to May 26, 2020, we prospectively identified hospitalized children with MIS-C (n = 10), symptomatic COVID-19 (n = 10), and KD (n = 5) and hospitalized controls (n = 4) at Children's Healthcare of Atlanta. With institutional review board approval, we obtained prospective and residual blood samples from these children and measured SARS-CoV-2 spike receptor-binding domain (RBD) immunoglobulin M and immunoglobulin G (IgG), full-length spike IgG, and nucleocapsid protein antibodies using quantitative enzyme-linked immunosorbent assays and SARS-CoV-2 neutralizing antibodies using live-virus focus-reduction neutralization assays. We statistically compared the log-transformed antibody titers among groups and performed linear regression analyses. RESULTS All children with MIS-C had high titers of SARS-CoV-2 RBD IgG antibodies, which correlated with full-length spike IgG antibodies (R 2 = 0.956; P < .001), nucleocapsid protein antibodies (R 2 = 0.846; P < .001), and neutralizing antibodies (R 2 = 0.667; P < .001). Children with MIS-C had significantly higher SARS-CoV-2 RBD IgG antibody titers (geometric mean titer 6800; 95% confidence interval 3495-13 231) than children with COVID-19 (geometric mean titer 626; 95% confidence interval 251-1563; P < .001), children with KD (geometric mean titer 124; 95% confidence interval 91-170; P < .001), and hospitalized controls (geometric mean titer 85; P < .001). All children with MIS-C also had detectable RBD immunoglobulin M antibodies, indicating recent SARS-CoV-2 infection. RBD IgG titers correlated with the erythrocyte sedimentation rate (R 2 = 0.512; P < .046) and with hospital (R 2 = 0.548; P = .014) and ICU lengths of stay (R 2 = 0.590; P = .010). CONCLUSIONS Quantitative SARS-CoV-2 serology may have a role in establishing the diagnosis of MIS-C, distinguishing it from similar clinical entities, and stratifying risk for adverse outcomes.
Collapse
Affiliation(s)
- Christina A Rostad
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia.,Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and School of Medicine, Emory University, Atlanta, Georgia; and
| | - Ann Chahroudi
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia.,Emory Vaccine Center and.,Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and School of Medicine, Emory University, Atlanta, Georgia; and
| | - Grace Mantus
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia.,Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and School of Medicine, Emory University, Atlanta, Georgia; and
| | - Stacey A Lapp
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia.,Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and School of Medicine, Emory University, Atlanta, Georgia; and
| | - Mehgan Teherani
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia.,Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and School of Medicine, Emory University, Atlanta, Georgia; and
| | - Lisa Macoy
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia.,Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and School of Medicine, Emory University, Atlanta, Georgia; and
| | - Keiko M Tarquinio
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Rajit K Basu
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Carol Kao
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia.,Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and School of Medicine, Emory University, Atlanta, Georgia; and
| | - W Matthew Linam
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia.,Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and School of Medicine, Emory University, Atlanta, Georgia; and
| | - Matthew G Zimmerman
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia.,Yerkes National Primate Research Center, Emory University, Atlanta, Georgia.,Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and School of Medicine, Emory University, Atlanta, Georgia; and
| | - Pei-Yong Shi
- Departments of Biochemistry and Molecular Biology and
| | - Vineet D Menachery
- Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas
| | - Matthew E Oster
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia
| | | | - Evan J Anderson
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia.,Department of Medicine, School of Medicine and.,Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and School of Medicine, Emory University, Atlanta, Georgia; and
| | - Mehul S Suthar
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia.,Emory Vaccine Center and.,Yerkes National Primate Research Center, Emory University, Atlanta, Georgia.,Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and School of Medicine, Emory University, Atlanta, Georgia; and
| | - Jens Wrammert
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia.,Emory Vaccine Center and.,Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and School of Medicine, Emory University, Atlanta, Georgia; and
| | - Preeti Jaggi
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia; .,Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and School of Medicine, Emory University, Atlanta, Georgia; and
| |
Collapse
|
33
|
Ladhani SN, Jeffery-Smith A, Patel M, Janarthanan R, Fok J, Crawley-Boevey E, Vusirikala A, Fernandez Ruiz De Olano E, Perez MS, Tang S, Dun-Campbell K, Evans EW, Bell A, Patel B, Amin-Chowdhury Z, Aiano F, Paranthaman K, Ma T, Saavedra-Campos M, Ellis J, Chand M, Brown K, Ramsay ME, Hopkins S, Shetty N, Chow JY, Gopal R, Zambon M. High prevalence of SARS-CoV-2 antibodies in care homes affected by COVID-19: Prospective cohort study, England. EClinicalMedicine 2020; 28:100597. [PMID: 33173854 PMCID: PMC7644437 DOI: 10.1016/j.eclinm.2020.100597] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/27/2020] [Accepted: 10/02/2020] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND We investigated six London care homes experiencing a COVID-19 outbreak and found high rates of SARS-CoV-2 infection among residents and staff. Here we report follow-up investigations including antibody testing in the same care homes five weeks later. METHODS Residents and staff in the initial investigation had a repeat nasal swab for SARS-CoV-2 RT-PCR and a blood test for SARS CoV-2 antibodies using ELISA based on SARS-CoV-2 native viral antigens derived from infected cells and virus neutralisation. FINDINGS Of the 518 residents and staff in the initial investigation, 186/241 (77.2%) surviving residents and 208/254 (81.9%) staff underwent serological testing. Almost all SARS-CoV-2 RT-PCR positive residents and staff were seropositive five weeks later, whether symptomatic (residents 35/35, 100%; staff, 22/22, 100%) or asymptomatic (residents 32/33, 97.0%; staff 21/22, 95.5%). Symptomatic but SARS-CoV-2 RT-PCR negative residents and staff also had high seropositivity rates (residents 23/27, 85.2%; staff 18/21, 85.7%), as did asymptomatic RT-PCR negative individuals (residents 61/91, 67.0%; staff 95/143, 66.4%). Neutralising antibody was detected in 118/132 (89.4%) seropositive individuals and was not associated with age or symptoms. Ten residents (10/79 re-tested, 12.7%) remained RT-PCR positive but with higher RT-PCR cycle threshold values; 7/10 had serological testing and all were seropositive. New infections were detected in three residents and one staff. INTERPRETATION RT-PCR provides a point prevalence of SARS-CoV-2 infection but significantly underestimates total exposure in outbreak settings. In care homes experiencing large COVID-19 outbreaks, most residents and staff had neutralising SARS-CoV-2 antibodies, which was not associated with age or symptoms. FUNDING PHE.
Collapse
Affiliation(s)
- Shamez N Ladhani
- Immunisation and Countermeasures Division, Public Health England, London, UK
- Paediatric Infectious Diseases Research Group, St. George's University of London, UK
- Corresponding author at: Immunisation and Countermeasures Division, Public Health England, 61 Colindale Avenue, London NW9 5EQ, UK.
| | | | - Monika Patel
- Virus Reference Department, Public Health England, London, UK
| | - Roshni Janarthanan
- London Health Protection Team, National Infection Service, Public Health England, London, UK
| | - Jonathan Fok
- London Health Protection Team, National Infection Service, Public Health England, London, UK
| | - Emma Crawley-Boevey
- London Health Protection Team, National Infection Service, Public Health England, London, UK
| | - Amoolya Vusirikala
- London Health Protection Team, National Infection Service, Public Health England, London, UK
| | | | - Marina Sanchez Perez
- London Health Protection Team, National Infection Service, Public Health England, London, UK
| | - Suzanne Tang
- London Health Protection Team, National Infection Service, Public Health England, London, UK
| | - Kate Dun-Campbell
- London Health Protection Team, National Infection Service, Public Health England, London, UK
| | - Edward Wynne- Evans
- London Health Protection Team, National Infection Service, Public Health England, London, UK
| | - Anita Bell
- London Health Protection Team, National Infection Service, Public Health England, London, UK
| | - Bharat Patel
- London Health Protection Team, National Infection Service, Public Health England, London, UK
| | | | - Felicity Aiano
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | | | - Thomas Ma
- Field Service, National Infection Service, Public Health England, UK
| | | | - Joanna Ellis
- Virus Reference Department, Public Health England, London, UK
| | - Meera Chand
- Antimicrobial Resistance and Hospital Acquired Infection Department, Public Health England, London, UK
| | - Kevin Brown
- Virus Reference Department, Public Health England, London, UK
| | - Mary E. Ramsay
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | - Susan Hopkins
- Antimicrobial Resistance and Hospital Acquired Infection Department, Public Health England, London, UK
| | - Nandini Shetty
- Virus Reference Department, Public Health England, London, UK
| | - J. Yimmy Chow
- London Health Protection Team, National Infection Service, Public Health England, London, UK
| | - Robin Gopal
- Virus Reference Department, Public Health England, London, UK
| | - Maria Zambon
- Virus Reference Department, Public Health England, London, UK
| |
Collapse
|
34
|
Akhand MRN, Azim KF, Hoque SF, Moli MA, Joy BD, Akter H, Afif IK, Ahmed N, Hasan M. Genome based evolutionary lineage of SARS-CoV-2 towards the development of novel chimeric vaccine. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2020; 85:104517. [PMID: 32882432 PMCID: PMC7462568 DOI: 10.1016/j.meegid.2020.104517] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/28/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023]
Abstract
The present study aimed to predict a novel chimeric vaccine by simultaneously targeting four major structural proteins via the establishment of ancestral relationship among different strains of coronaviruses. Conserved regions from the homologous protein sets of spike glycoprotein, membrane protein, envelope protein and nucleocapsid protein were identified through multiple sequence alignment. The phylogeny analyses of whole genome stated that four proteins reflected the close ancestral relation of SARS-CoV-2 to SARS-COV-1 and bat coronavirus. Numerous immunogenic epitopes (both T cell and B cell) were generated from the common fragments which were further ranked on the basis of antigenicity, transmembrane topology, conservancy level, toxicity and allergenicity pattern and population coverage analysis. Top putative epitopes were combined with appropriate adjuvants and linkers to construct a novel multiepitope subunit vaccine against COVID-19. The designed constructs were characterized based on physicochemical properties, allergenicity, antigenicity and solubility which revealed the superiority of construct V3 in terms safety and efficacy. Essential molecular dynamics and normal mode analysis confirmed minimal deformability of the refined model at molecular level. In addition, disulfide engineering was investigated to accelerate the stability of the protein. Molecular docking study ensured high binding affinity between construct V3 and HLA cells, as well as with different host receptors. Microbial expression and translational efficacy of the constructs were checked using pET28a(+) vector of E. coli strain K12. However, the in vivo and in vitro validation of suggested vaccine molecule might be ensured with wet lab trials using model animals for the implementation of the presented data.
Collapse
Affiliation(s)
- Mst Rubaiat Nazneen Akhand
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh,Department of Biochemistry and Chemistry, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Kazi Faizul Azim
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh,Department of Microbial Biotechnology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Syeda Farjana Hoque
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh,Department of Pharmaceuticals and Industrial Biotechnology, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Mahmuda Akther Moli
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh,Department of Pharmaceuticals and Industrial Biotechnology, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Bijit Das Joy
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh,Department of Biochemistry and Chemistry, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Hafsa Akter
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Ibrahim Khalil Afif
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Nadim Ahmed
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Mahmudul Hasan
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh; Department of Pharmaceuticals and Industrial Biotechnology, Sylhet Agricultural University, Sylhet 3100, Bangladesh.
| |
Collapse
|
35
|
Huang AT, Garcia-Carreras B, Hitchings MDT, Yang B, Katzelnick LC, Rattigan SM, Borgert BA, Moreno CA, Solomon BD, Trimmer-Smith L, Etienne V, Rodriguez-Barraquer I, Lessler J, Salje H, Burke DS, Wesolowski A, Cummings DAT. A systematic review of antibody mediated immunity to coronaviruses: kinetics, correlates of protection, and association with severity. Nat Commun 2020; 11:4704. [PMID: 32943637 PMCID: PMC7499300 DOI: 10.1038/s41467-020-18450-4] [Citation(s) in RCA: 636] [Impact Index Per Article: 127.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/18/2020] [Indexed: 01/05/2023] Open
Abstract
Many public health responses and modeled scenarios for COVID-19 outbreaks caused by SARS-CoV-2 assume that infection results in an immune response that protects individuals from future infections or illness for some amount of time. The presence or absence of protective immunity due to infection or vaccination (when available) will affect future transmission and illness severity. Here, we review the scientific literature on antibody immunity to coronaviruses, including SARS-CoV-2 as well as the related SARS-CoV, MERS-CoV and endemic human coronaviruses (HCoVs). We reviewed 2,452 abstracts and identified 491 manuscripts relevant to 5 areas of focus: 1) antibody kinetics, 2) correlates of protection, 3) immunopathogenesis, 4) antigenic diversity and cross-reactivity, and 5) population seroprevalence. While further studies of SARS-CoV-2 are necessary to determine immune responses, evidence from other coronaviruses can provide clues and guide future research.
Collapse
Affiliation(s)
- Angkana T Huang
- Department of Biology, University of Florida, Gainesville, FL, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Bernardo Garcia-Carreras
- Department of Biology, University of Florida, Gainesville, FL, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Matt D T Hitchings
- Department of Biology, University of Florida, Gainesville, FL, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Bingyi Yang
- Department of Biology, University of Florida, Gainesville, FL, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Leah C Katzelnick
- Department of Biology, University of Florida, Gainesville, FL, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Susan M Rattigan
- Department of Biology, University of Florida, Gainesville, FL, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Brooke A Borgert
- Department of Biology, University of Florida, Gainesville, FL, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Carlos A Moreno
- Department of Biology, University of Florida, Gainesville, FL, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Benjamin D Solomon
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Luke Trimmer-Smith
- Department of Biology, University of Florida, Gainesville, FL, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Veronique Etienne
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
- Department of Comparative, Diagnostic & Population Medicine, University of Florida, Gainesville, FL, USA
| | | | - Justin Lessler
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Henrik Salje
- Department of Biology, University of Florida, Gainesville, FL, USA
- Department of Genetics, University of Cambridge, Cambridge, UK
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, Paris, France
| | - Donald S Burke
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amy Wesolowski
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Derek A T Cummings
- Department of Biology, University of Florida, Gainesville, FL, USA.
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
36
|
Zaichuk TA, Nechipurenko YD, Adzhubey AA, Onikienko SB, Chereshnev VA, Zainutdinov SS, Kochneva GV, Netesov SV, Matveeva OV. The Challenges of Vaccine Development against Betacoronaviruses: Antibody Dependent Enhancement and Sendai Virus as a Possible Vaccine Vector. Mol Biol 2020; 54:812-826. [PMID: 32921819 PMCID: PMC7473411 DOI: 10.1134/s0026893320060151] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/21/2022]
Abstract
To design an effective and safe vaccine against betacoronaviruses, it is necessary to use their evolutionarily conservative antigenic determinants that will elicit the combination of strong humoral and cell-mediated immune responses. Targeting such determinants minimizes the risk of antibody-dependent enhancement of viral infection. This phenomenon was observed in animal trials of experimental vaccines against SARS-CoV-1 and MERS-CoV that were developed based on inactivated coronavirus or vector constructs expressing the spike protein (S) of the virion. The substitution and glycosylation of certain amino acids in the antigenic determinants of the S-protein, as well as its conformational changes, can lead to the same effect in a new experimental vaccine against SARS-CoV-2. Using more conservative structural and accessory viral proteins for the vaccine antigenic determinants will help to avoid this problem. This review outlines approaches for developing vaccines against the new SARS-CoV-2 coronavirus that are based on non-pathogenic viral vectors. For efficient prevention of infections caused by respiratory pathogens the ability of the vaccine to stimulate mucosal immunity in the respiratory tract is important. Such a vaccine can be developed using non-pathogenic Sendai virus vector, since it can be administered intranasally and induce a mucosal immune response that strengthens the antiviral barrier in the respiratory tract and provides reliable protection against infection.
Collapse
Affiliation(s)
| | - Y D Nechipurenko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - A A Adzhubey
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia.,George Washington University, 20052 Washington, DC USA
| | - S B Onikienko
- Department of Military Field Therapy, Kirov Military Medical Academy, 194044 St. Petersburg, Russia
| | - V A Chereshnev
- Institute of Immunology and Physiology, 620049 Yekaterinburg, Russia
| | - S S Zainutdinov
- State Research Center of Virology and Biotechnology "Vector,", 630559 Koltsovo, Russia
| | - G V Kochneva
- State Research Center of Virology and Biotechnology "Vector,", 630559 Koltsovo, Russia
| | - S V Netesov
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - O V Matveeva
- Sendai Viralytics, 117261 Acton, MA USA.,Biopolymer Design, 117281 Acton, MA USA
| |
Collapse
|
37
|
Karthik K, Senthilkumar TMA, Udhayavel S, Raj GD. Role of antibody-dependent enhancement (ADE) in the virulence of SARS-CoV-2 and its mitigation strategies for the development of vaccines and immunotherapies to counter COVID-19. Hum Vaccin Immunother 2020; 16:3055-3060. [PMID: 32845733 PMCID: PMC7484565 DOI: 10.1080/21645515.2020.1796425] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Coronavirus disease-2019 (COVID-19) pandemic has become a global threat and death tolls are increasing worldwide. The SARS-CoV-2 though shares similarities with SARS-CoV and MERS-CoV, immunopathology of the novel virus is not understood properly. Previous reports from SARS and MERS-CoV documents that preexisting, non-neutralizing or poorly neutralizing antibodies developed as a result of vaccine or infection enhance subsequent infection, a phenomenon called as antibody-dependent enhancement (ADE). Since immunotherapy has been implicated for COVID-19 treatment and vaccine is under development, due consideration has to be provided on ADE to prevent untoward reactions. ADE mitigation strategies like the development of vaccine or immunotherapeutics targeting receptor binding motif can be designed to minimize ADE of SARS-CoV-2 since full-length protein-based approach can lead to ADE as reported in MERS-CoV. The present mini-review aims to address the phenomenon of ADE of SARS-CoV-2 through the lessons learned from SARS-CoV and MERS-CoV and ways to mitigate them so as to develop better vaccines and immunotherapeutics against SARS-CoV-2.
Collapse
Affiliation(s)
- Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University , Chennai, India
| | | | - Shanmugasundaram Udhayavel
- Poultry Disease Diagnosis and Surveillance Laboratory, Tamil Nadu Veterinary and Animal Sciences University , Chennai, India
| | - Gopal Dhinakar Raj
- Centre for Animal Health Studies, Tamil Nadu Veterinary and Animal Sciences University , Chennai, India
| |
Collapse
|
38
|
Melenotte C, Silvin A, Goubet AG, Lahmar I, Dubuisson A, Zumla A, Raoult D, Merad M, Gachot B, Hénon C, Solary E, Fontenay M, André F, Maeurer M, Ippolito G, Piacentini M, Wang FS, Ginhoux F, Marabelle A, Kroemer G, Derosa L, Zitvogel L. Immune responses during COVID-19 infection. Oncoimmunology 2020; 9:1807836. [PMID: 32939324 PMCID: PMC7480812 DOI: 10.1080/2162402x.2020.1807836] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 02/09/2023] Open
Abstract
Over the past 16 years, three coronaviruses (CoVs), severe acute respiratory syndrome CoV (SARS-CoV) in 2002, Middle East respiratory syndrome CoV (MERS-CoV) in 2012 and 2015, and SARS-CoV-2 in 2020, have been causing severe and fatal human epidemics. The unpredictability of coronavirus disease-19 (COVID-19) poses a major burden on health care and economic systems across the world. This is caused by the paucity of in-depth knowledge of the risk factors for severe COVID-19, insufficient diagnostic tools for the detection of SARS-CoV-2, as well as the absence of specific and effective drug treatments. While protective humoral and cellular immune responses are usually mounted against these betacoronaviruses, immune responses to SARS-CoV2 sometimes derail towards inflammatory tissue damage, leading to rapid admissions to intensive care units. The lack of knowledge on mechanisms that tilt the balance between these two opposite outcomes poses major threats to many ongoing clinical trials dealing with immunostimulatory or immunoregulatory therapeutics. This review will discuss innate and cognate immune responses underlying protective or deleterious immune reactions against these pathogenic coronaviruses.
Collapse
Affiliation(s)
- Cléa Melenotte
- Immunology, Gustave Roussy, Villejuif, France
- Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Infectious Diseases, Aix-Marseille Université, IRD, APHM, MEPHI, Marseille, France
- Infectious Diseases, IHU-Méditerranée Infection, Marseille, France
| | | | - Anne-Gaëlle Goubet
- Immunology, Gustave Roussy, Villejuif, France
- Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Immunology, Institut National de la Santé Et de la Recherche Médicale (INSERM), U1015 Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Imran Lahmar
- Immunology, Gustave Roussy, Villejuif, France
- Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Immunology, Institut National de la Santé Et de la Recherche Médicale (INSERM), U1015 Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Agathe Dubuisson
- Immunology, Gustave Roussy, Villejuif, France
- Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Immunology, Institut National de la Santé Et de la Recherche Médicale (INSERM), U1015 Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Alimuddin Zumla
- Department of Infection, Division of Infection and Immunity, University College London, National Institute for Health Research Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, UK
| | - Didier Raoult
- Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Infectious Diseases, Aix-Marseille Université, IRD, APHM, MEPHI, Marseille, France
| | - Mansouria Merad
- Service de Urgences et de Permanence des Soins, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | | | | | - Eric Solary
- Immunology, Gustave Roussy, Villejuif, France
| | - Michaela Fontenay
- INSERM U1016, Centre National Recherche Scientifique (CNRS) UMR8104, Institut Cochin, Université de Paris, Paris, France
| | | | - Markus Maeurer
- Immunosurgery, Immunotherapy Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
- Med Clinic, University of Mainz, Mayence, Germany
| | - Giuseppe Ippolito
- Dipartimento di Epidemiologia Ricerca Pre-Clinica e Diagnostica Avanzata, National Institute for Infectious Diseases “Lazzaro Spallanzani” I.R.C.C.S., Rome, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
- Infectious Diseases Department, National Institute for Infectious Disease IRCCS “Lazzaro Spallanzani”, Rome, Italy
| | - Fu-Sheng Wang
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Aurélien Marabelle
- Infectious Diseases, Aix-Marseille Université, IRD, APHM, MEPHI, Marseille, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Pôle de Biologie,Pathologie – PUI – Hygiène, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Lisa Derosa
- Immunology, Gustave Roussy, Villejuif, France
- Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Immunology, Institut National de la Santé Et de la Recherche Médicale (INSERM), U1015 Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Laurence Zitvogel
- Immunology, Gustave Roussy, Villejuif, France
- Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Immunology, Institut National de la Santé Et de la Recherche Médicale (INSERM), U1015 Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| |
Collapse
|
39
|
Santos IDA, Grosche VR, Bergamini FRG, Sabino-Silva R, Jardim ACG. Antivirals Against Coronaviruses: Candidate Drugs for SARS-CoV-2 Treatment? Front Microbiol 2020; 11:1818. [PMID: 32903349 PMCID: PMC7438404 DOI: 10.3389/fmicb.2020.01818] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/10/2020] [Indexed: 01/08/2023] Open
Abstract
Coronaviruses (CoVs) are a group of viruses from the family Coronaviridae that can infect humans and animals, causing mild to severe diseases. The ongoing pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) represents a global threat, urging the development of new therapeutic strategies. Here we present a selection of relevant compounds that have been described from 2005 until now as having in vitro and/or in vivo antiviral activities against human and/or animal CoVs. We also present compounds that have reached clinical trials as well as further discussing the potentiality of other molecules for application in (re)emergent CoVs outbreaks. Finally, through rationalization of the data presented herein, we wish to encourage further research encompassing these compounds as potential SARS-CoV-2 drug candidates.
Collapse
Affiliation(s)
- Igor de Andrade Santos
- Laboratory of Virology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Victória Riquena Grosche
- Laboratory of Virology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
- Institute of Biosciences, Language and Exact Sciences, São Paulo State University, São José do Rio Preto, Brazil
| | | | - Robinson Sabino-Silva
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Ana Carolina Gomes Jardim
- Laboratory of Virology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
- Institute of Biosciences, Language and Exact Sciences, São Paulo State University, São José do Rio Preto, Brazil
| |
Collapse
|
40
|
Ayouba A, Thaurignac G, Morquin D, Tuaillon E, Raulino R, Nkuba A, Lacroix A, Vidal N, Foulongne V, Le Moing V, Reynes J, Delaporte E, Peeters M. Multiplex detection and dynamics of IgG antibodies to SARS-CoV2 and the highly pathogenic human coronaviruses SARS-CoV and MERS-CoV. J Clin Virol 2020; 129:104521. [PMID: 32623350 PMCID: PMC7308014 DOI: 10.1016/j.jcv.2020.104521] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/15/2020] [Accepted: 06/21/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Knowledge of the COVID-19 epidemic extent and the level of herd immunity is urgently needed to help manage this pandemic. METHODS We used a panel of 167 samples (77 pre-epidemic and 90 COVID-19 seroconverters) and SARS-CoV1, SARS-CoV2 and MERS-CoV Spike and/or Nucleopcapsid (NC) proteins to develop a high throughput multiplex screening assay to detect IgG antibodies in human plasma. Assay performances were determined by ROC curves analysis. A subset of the COVID-19+ samples (n = 36) were also tested by a commercial NC-based ELISA test and the results compared with those of the novel assay. RESULTS On samples collected ≥14 days after symptoms onset, the accuracy of the assay is 100 % (95 % CI: 100-100) for the Spike antigen and 99.9 % (95 % CI:99.7-100) for NC. By logistic regression, we estimated that 50 % of the patients have seroconverted at 5.7 ± 1.6; 5.7 ± 1.8 and 7.9 ± 1.0 days after symptoms onset against Spike, NC or both antigens, respectively and all have seroconverted two weeks after symptoms onset. IgG titration in a subset of samples showed that early phase samples present lower IgG titers than those from later phase. IgG to SARS-CoV2 NC cross-reacted at 100 % with SARS-CoV1 NC. Twenty-nine of the 36 (80.5 %) samples tested were positive by the commercial ELISA while 31/36 (86.1 %) were positive by the novel assay. CONCLUSIONS Our assay is highly sensitive and specific for the detection of IgG antibodies to SARS-CoV2 proteins, suitable for high throughput epidemiological surveys. The novel assay is more sensitive than a commercial ELISA.
Collapse
Affiliation(s)
- Ahidjo Ayouba
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175, Institut de Recherche pour le Développement et Université de Montpellier, France.
| | - Guillaume Thaurignac
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175, Institut de Recherche pour le Développement et Université de Montpellier, France
| | - David Morquin
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175, Institut de Recherche pour le Développement et Université de Montpellier, France; Département de Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Edouard Tuaillon
- Pathogenesis and Control of Chronic Infections, University of Montpellier, INSERM, EFS, CHU Montpellier, Montpellier, France
| | - Raisa Raulino
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175, Institut de Recherche pour le Développement et Université de Montpellier, France
| | - Antoine Nkuba
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175, Institut de Recherche pour le Développement et Université de Montpellier, France
| | - Audrey Lacroix
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175, Institut de Recherche pour le Développement et Université de Montpellier, France
| | - Nicole Vidal
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175, Institut de Recherche pour le Développement et Université de Montpellier, France
| | - Vincent Foulongne
- Département de bacteriologie-virologie, CHU de Montpellier, 34295 Montpellier, France
| | - Vincent Le Moing
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175, Institut de Recherche pour le Développement et Université de Montpellier, France; Département de Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Jacques Reynes
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175, Institut de Recherche pour le Développement et Université de Montpellier, France; Département de Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Eric Delaporte
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175, Institut de Recherche pour le Développement et Université de Montpellier, France; Département de Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Martine Peeters
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175, Institut de Recherche pour le Développement et Université de Montpellier, France
| |
Collapse
|
41
|
Harvala H, Mehew J, Robb ML, Ijaz S, Dicks S, Patel M, Watkins N, Simmonds P, Brooks T, Johnson R, Gopal R, Roberts DJ, Zambon M. Convalescent plasma treatment for SARS-CoV-2 infection: analysis of the first 436 donors in England, 22 April to 12 May 2020. Euro Surveill 2020; 25:2001260. [PMID: 32700670 PMCID: PMC7376844 DOI: 10.2807/1560-7917.es.2020.25.28.2001260] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/16/2020] [Indexed: 12/29/2022] Open
Abstract
Serological reactivity was analysed in plasma from 436 individuals with a history of disease compatible with COVID-19, including 256 who had been laboratory-confirmed with SARS-CoV-2 infection. Over 99% of laboratory-confirmed cases developed a measurable antibody response (254/256) and 88% harboured neutralising antibodies (226/256). Antibody levels declined over 3 months following diagnosis, emphasising the importance of the timing of convalescent plasma collections. Binding antibody measurements can inform selection of convalescent plasma donors with high neutralising antibody levels.
Collapse
Affiliation(s)
- Heli Harvala
- Microbiology Services, NHS Blood and Transplant, London, United Kingdom
| | - Jennifer Mehew
- Statistics and Clinical Studies, NHS Blood and Transplant, Bristol, United Kingdom
| | - Matthew L Robb
- Statistics and Clinical Studies, NHS Blood and Transplant, Bristol, United Kingdom
| | - Samreen Ijaz
- Virology Reference Department, National Infection Service, Public Health England, Colindale Avenue, London, United Kingdom
| | - Steven Dicks
- Microbiology Services, NHS Blood and Transplant, London, United Kingdom
- Virology Reference Department, National Infection Service, Public Health England, Colindale Avenue, London, United Kingdom
| | - Monika Patel
- High Containment Microbiology, National Infection Service, Public Health England, Colindale Avenue, London, United Kingdom
| | | | - Peter Simmonds
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Tim Brooks
- Rare & Imported Pathogens Laboratory, Public Health England, Porton Down, United Kingdom
| | - Rachel Johnson
- Statistics and Clinical Studies, NHS Blood and Transplant, Bristol, United Kingdom
| | - Robin Gopal
- High Containment Microbiology, National Infection Service, Public Health England, Colindale Avenue, London, United Kingdom
| | - David J Roberts
- NHS Blood and Transplant, Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Radcliffe Department of Medicine and BRC Haematology Theme, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Maria Zambon
- Virology Reference Department, National Infection Service, Public Health England, Colindale Avenue, London, United Kingdom
| |
Collapse
|
42
|
康 庄, 唐 梅. [Progress and analysis on the development of 2019-nCoV vaccine]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2020; 37:373-379. [PMID: 32597077 PMCID: PMC10319570 DOI: 10.7507/1001-5515.202004025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Indexed: 11/03/2022]
Abstract
As the COVID-19 pandemic is intensifying globally, more and more people are pinning their hopes on the development of vaccines. At present, there are many research teams who have adopted different vaccine technology routes to develop 2019-nCoV vaccines. This article reviews and analyzes the current development and research status of 2019-nCoV vaccines in different routes, and explores their possible development in the future.
Collapse
Affiliation(s)
- 庄 康
- 乐山师范学院 生命科学学院(四川乐山 614000)College of Life Science, Leshan Normal University, Leshan, Sichuan 614000, P.R.China
- 成都生物制品研究所有限责任公司 生物技术研究室(成都 610000)Department of Biotechnology, Chengdu Institute of Biological Products, Chengdu 610000, P.R.China
| | - 梅 唐
- 乐山师范学院 生命科学学院(四川乐山 614000)College of Life Science, Leshan Normal University, Leshan, Sichuan 614000, P.R.China
| |
Collapse
|
43
|
Hassan MM, Hussain MA, Kambal S, Elshikh AA, Gendeel OR, Ahmed SA, Altayeb RA, Muhajir AM, Mohamed SB. NeoCoV Is Closer to MERS-CoV than SARS-CoV. Infect Dis (Lond) 2020; 13:1178633720930711. [PMID: 32595278 PMCID: PMC7298434 DOI: 10.1177/1178633720930711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/07/2020] [Indexed: 01/12/2023] Open
Abstract
Recently, Coronavirus has been given considerable attention from the biomedical
community based on the emergence and isolation of a deadly coronavirus infecting
human. To understand the behavior of the newly emerging MERS-CoV requires
knowledge at different levels (epidemiologic, antigenic, and pathogenic), and
this knowledge can be generated from the most related viruses. In this study, we
aimed to compare between 3 species of Coronavirus, namely Middle East
Respiratory Syndrome (MERS-CoV), Severe Acute Respiratory Syndrome (SARS-CoV),
and NeoCoV regarding whole genomes and 6 similar proteins (E, M, N, S, ORF1a,
and ORF1ab) using different bioinformatics tools to provide a better
understanding of the relationship between the 3 viruses at the nucleotide and
amino acids levels. All sequences have been retrieved from National Center for
Biotechnology Information (NCBI). Regards to target genomes’ phylogenetic
analysis showed that MERS and SARS-CoVs were closer to each other compared with
NeoCoV, and the last has the longest relative time. We found that all
phylogenetic methods in addition to all parameters (physical and chemical
properties of amino acids such as the number of amino acid, molecular weight,
atomic composition, theoretical pI, and structural formula) indicated that
NeoCoV proteins were the most related to MERS-CoV one. All phylogenetic trees
(by both maximum-likelihood and neighbor-joining methods) indicated that NeoCoV
proteins have less evolutionary changes except for ORF1a by just
maximum-likelihood method. Our results indicated high similarity between viral
structural proteins which are responsible for viral infectivity; therefore, we
expect that NeoCoV sooner may appear in human-related infection.
Collapse
Affiliation(s)
- Mohamed M Hassan
- Bioinformatics and Biostatistics Department, National University Biomedical Research Institute, National University, Khartoum, Sudan
| | - Mohamed A Hussain
- Department of Pharmaceutical Microbiology, International University of Africa, Khartoum, Sudan
| | - Sumaya Kambal
- Bioinformatics and Biostatistics Department, National University Biomedical Research Institute, National University, Khartoum, Sudan
| | - Ahmed A Elshikh
- Department of Microbiology, Faculty of Pure and Applied Sciences, International University of Africa, Khartoum, Sudan
| | - Osama R Gendeel
- Faculty of Science and Technology, Omdurman Islamic University, Omdurman, Sudan
| | - Siddig A Ahmed
- Faculty of Science and Technology, Omdurman Islamic University, Omdurman, Sudan
| | - Rami A Altayeb
- Faculty of Science and Technology, Omdurman Islamic University, Omdurman, Sudan
| | | | - Sofia B Mohamed
- Bioinformatics and Biostatistics Department, National University Biomedical Research Institute, National University, Khartoum, Sudan
| |
Collapse
|
44
|
Bundschuh C, Egger M, Wiesinger K, Gabriel C, Clodi M, Mueller T, Dieplinger B. Evaluation of the EDI enzyme linked immunosorbent assays for the detection of SARS-CoV-2 IgM and IgG antibodies in human plasma. Clin Chim Acta 2020; 509:79-82. [PMID: 32526218 PMCID: PMC7278646 DOI: 10.1016/j.cca.2020.05.047] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 01/24/2023]
Abstract
Evaluation of EDITM ELISAs for detection of SARS-CoV-2 IgM and IgG in human plasma. Antibodies were measured in 64 SARS-CoV-2 RT-PCR confirmed COVID-19 patients. with serial blood samples collected at different time points from symptom onset. In 200 healthy blood donors, and in 256 intensive care unit patients. High “true” vs. low “false” positivity rates for the SARS-CoV-2 IgM and IgG ELISAs.
Background Besides SARS-CoV-2 RT-PCR testing, serological testing is emerging as additional option in COVID-19 diagnostics. Aim of this study was to evaluate novel immunoassays for detection of SARS-CoV-2 antibodies in human plasma. Methods Using EDITM Novel Coronavirus COVID-19 Enzyme Linked Immunosorbent Assays (ELISAs), we measured SARS-CoV-2 IgM and IgG antibodies in 64 SARS-CoV-2 RT-PCR confirmed COVID-19 patients with serial blood samples (n = 104) collected at different time points from symptom onset. Blood samples from 200 healthy blood donors and 256 intensive care unit (ICU) patients collected before the COVID-19 outbreak were also used. Results The positivity rates in the COVID-19 patients were 5.9% for IgM and 2.9% for IgG ≤ 5 days after symptom onset; Between day 5 and day 10 the positivity rates were 37.1% for IgM and 37.1% for IgG and rose to 76.4% for IgM and 82.4% for IgG after > 10–15 days. After 15–22 days the “true” positivity rates were 94.4% for IgM and 100% for IgG. The “false” positivity rates were 0.5% for IgM and 1.0% for IgG in the healthy blood donors, 1.6% for IgM and 1.2% for IgG in ICU patients. Conclusions This study shows high “true” vs. low “false” positivity rates for the EDITM SARS-CoV-2 IgM and IgG ELISAs.
Collapse
Affiliation(s)
- Christian Bundschuh
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder Linz and Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria
| | - Margot Egger
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder Linz and Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria
| | - Kurt Wiesinger
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder Linz and Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria
| | - Christian Gabriel
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology Vienna, Vienna, Austria
| | - Martin Clodi
- Department of Internal Medicine, Konventhospital Barmherzige Brueder Linz, Linz, Austria
| | - Thomas Mueller
- Department of Clinical Pathology, Hospital of Bolzano, Bolzano, Italy
| | - Benjamin Dieplinger
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder Linz and Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria.
| |
Collapse
|
45
|
Padron-Regalado E. Vaccines for SARS-CoV-2: Lessons from Other Coronavirus Strains. Infect Dis Ther 2020; 9:255-274. [PMID: 32328406 PMCID: PMC7177048 DOI: 10.1007/s40121-020-00300-x] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Indexed: 12/13/2022] Open
Abstract
The emergence of the strain of coronavirus SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) and its impact on global health have made imperative the development of effective and safe vaccines for this lethal strain. SARS-CoV-2 now adds to the list of coronavirus diseases that have threatened global health, along with the SARS (severe acute respiratory syndrome) and MERS (Middle East respiratory syndrome) coronaviruses that emerged in 2002/2003 and 2012, respectively. As of April 2020, no vaccine is commercially available for these coronavirus strains. Nevertheless, the knowledge obtained from the vaccine development efforts for MERS and SARS can be of high value for COVID-19 (coronavirus disease 2019). Here, we review the past and ongoing vaccine development efforts for clinically relevant coronavirus strains with the intention that this information helps in the development of effective and safe vaccines for COVID-19. In addition, information from naturally exposed individuals and animal models to coronavirus strains is described for the same purpose of helping into the development of effective vaccines against COVID-19.
Collapse
|
46
|
de Assis RR, Jain A, Nakajima R, Jasinskas A, Felgner J, Obiero JM, Adenaiye O, Tai S, Hong F, Norris PJ, Stone M, Simmons G, Bagri A, Schreiber M, Buser A, Holbro A, Battegay M, Hosimer P, Noesen C, Milton DK, Davies DH, Contestable P, Corash LM, Busch MP, Felgner PL, Khan S. Analysis of SARS-CoV-2 Antibodies in COVID-19 Convalescent Blood using a Coronavirus Antigen Microarray. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32511302 PMCID: PMC7217240 DOI: 10.1101/2020.04.15.043364] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The current practice for diagnosis of COVID-19, based on SARS-CoV-2 PCR testing of pharyngeal or respiratory specimens in a symptomatic patient at high epidemiologic risk, likely underestimates the true prevalence of infection. Serologic methods can more accurately estimate the disease burden by detecting infections missed by the limited testing performed to date. Here, we describe the validation of a coronavirus antigen microarray containing immunologically significant antigens from SARS-CoV-2, in addition to SARS-CoV, MERS-CoV, common human coronavirus strains, and other common respiratory viruses. A comparison of antibody profiles detected on the array from control sera collected prior to the SARS-CoV-2 pandemic versus convalescent blood specimens from virologically confirmed COVID-19 cases demonstrates near complete discrimination of these two groups, with improved performance from use of antigen combinations that include both spike protein and nucleoprotein. This array can be used as a diagnostic tool, as an epidemiologic tool to more accurately estimate the disease burden of COVID-19, and as a research tool to correlate antibody responses with clinical outcomes.
Collapse
Affiliation(s)
- Rafael R de Assis
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA
| | - Aarti Jain
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA
| | - Rie Nakajima
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA
| | - Algis Jasinskas
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA
| | - Jiin Felgner
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA
| | - Joshua M Obiero
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA
| | - Oluwasanmi Adenaiye
- Institute for Applied Environmental Health, School of Public Health, University of Maryland, College Park, MD
| | - Sheldon Tai
- Institute for Applied Environmental Health, School of Public Health, University of Maryland, College Park, MD
| | - Filbert Hong
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA
| | - Philip J Norris
- Vitalant Research Institute, San Francisco, CA.,Department of Laboratory Medicine, University of California, San Francisco, CA
| | - Mars Stone
- Vitalant Research Institute, San Francisco, CA.,Department of Laboratory Medicine, University of California, San Francisco, CA
| | - Graham Simmons
- Vitalant Research Institute, San Francisco, CA.,Department of Laboratory Medicine, University of California, San Francisco, CA
| | | | - Martin Schreiber
- Department of Surgery, Oregon Health & Science University, Portland, OR
| | - Andreas Buser
- Division of Infectious Diseases & Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Andreas Holbro
- Division of Infectious Diseases & Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Manuel Battegay
- Division of Infectious Diseases & Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | | | | | - Donald K Milton
- Institute for Applied Environmental Health, School of Public Health, University of Maryland, College Park, MD
| | | | - D Huw Davies
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA
| | | | | | - Michael P Busch
- Vitalant Research Institute, San Francisco, CA.,Department of Laboratory Medicine, University of California, San Francisco, CA
| | - Philip L Felgner
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA
| | - Saahir Khan
- Division of Infectious Diseases, Department of Medicine, University of California Irvine Health, Orange, CA
| |
Collapse
|
47
|
Tse LV, Meganck RM, Graham RL, Baric RS. The Current and Future State of Vaccines, Antivirals and Gene Therapies Against Emerging Coronaviruses. Front Microbiol 2020; 11:658. [PMID: 32390971 PMCID: PMC7193113 DOI: 10.3389/fmicb.2020.00658] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/23/2020] [Indexed: 12/31/2022] Open
Abstract
Emerging coronaviruses (CoV) are constant global public health threats to society. Multiple ongoing clinical trials for vaccines and antivirals against CoVs showcase the availability of medical interventions to both prevent and treat the future emergence of highly pathogenic CoVs in human. However, given the diverse nature of CoVs and our close interactions with wild, domestic and companion animals, the next epidemic zoonotic CoV could resist the existing vaccines and antivirals developed, which are primarily focused on Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and Middle East Respiratory Syndrome Coronavirus (MERS CoV). In late 2019, the novel CoV (SARS-CoV-2) emerged in Wuhan, China, causing global public health concern. In this review, we will summarize the key advancements of current vaccines and antivirals against SARS-CoV and MERS-CoV as well as discuss the challenge and opportunity in the current SARS-CoV-2 crisis. At the end, we advocate the development of a "plug-and-play" platform technologies that could allow quick manufacturing and administration of broad-spectrum countermeasures in an outbreak setting. We will discuss the potential of AAV-based gene therapy technology for in vivo therapeutic antibody delivery to combat SARS-CoV-2 outbreak and the future emergence of severe CoVs.
Collapse
Affiliation(s)
- Longping V. Tse
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rita M. Meganck
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rachel L. Graham
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ralph S. Baric
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
48
|
Huang AT, Garcia-Carreras B, Hitchings MD, Yang B, Katzelnick LC, Rattigan SM, Borgert BA, Moreno CA, Solomon BD, Rodriguez-Barraquer I, Lessler J, Salje H, Burke D, Wesolowski A, Cummings DA. A systematic review of antibody mediated immunity to coronaviruses: antibody kinetics, correlates of protection, and association of antibody responses with severity of disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.04.14.20065771. [PMID: 32511434 PMCID: PMC7217088 DOI: 10.1101/2020.04.14.20065771] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
The duration and nature of immunity generated in response to SARS-CoV-2 infection is unknown. Many public health responses and modeled scenarios for COVID-19 outbreaks caused by SARSCoV-2 assume that infection results in an immune response that protects individuals from future infections or illness for some amount of time. The timescale of protection is a critical determinant of the future impact of the pathogen. The presence or absence of protective immunity due to infection or vaccination (when available) will affect future transmission and illness severity. The dynamics of immunity and nature of protection are relevant to discussions surrounding therapeutic use of convalescent sera as well as efforts to identify individuals with protective immunity. Here, we review the scientific literature on antibody immunity to coronaviruses, including SARS-CoV-2 as well as the related SARS-CoV-1, MERS-CoV and human endemic coronaviruses (HCoVs). We reviewed 1281 abstracts and identified 322 manuscripts relevant to 5 areas of focus: 1) antibody kinetics, 2) correlates of protection, 3) immunopathogenesis, 4) antigenic diversity and cross-reactivity, and 5) population seroprevalence. While studies of SARS-CoV-2 are necessary to determine immune responses to it, evidence from other coronaviruses can provide clues and guide future research.
Collapse
Affiliation(s)
- Angkana T. Huang
- Department of Biology, University of Florida, USA
- Emerging Pathogens Institute, University of Florida, USA
| | - Bernardo Garcia-Carreras
- Department of Biology, University of Florida, USA
- Emerging Pathogens Institute, University of Florida, USA
| | - Matt D.T. Hitchings
- Department of Biology, University of Florida, USA
- Emerging Pathogens Institute, University of Florida, USA
| | - Bingyi Yang
- Department of Biology, University of Florida, USA
- Emerging Pathogens Institute, University of Florida, USA
| | - Leah C. Katzelnick
- Department of Biology, University of Florida, USA
- Emerging Pathogens Institute, University of Florida, USA
| | - Susan M. Rattigan
- Department of Biology, University of Florida, USA
- Emerging Pathogens Institute, University of Florida, USA
| | - Brooke A. Borgert
- Department of Biology, University of Florida, USA
- Emerging Pathogens Institute, University of Florida, USA
| | - Carlos A. Moreno
- Department of Biology, University of Florida, USA
- Emerging Pathogens Institute, University of Florida, USA
| | - Benjamin D. Solomon
- National Human Genome Research Institute, National Institutes of Health, USA
| | | | - Justin Lessler
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, USA
| | - Henrik Salje
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Donald Burke
- Department of Epidemiology, University of Pittsburgh, USA
| | - Amy Wesolowski
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, USA
| | - Derek A.T. Cummings
- Department of Biology, University of Florida, USA
- Emerging Pathogens Institute, University of Florida, USA
| |
Collapse
|
49
|
Khan S, Nakajima R, Jain A, de Assis RR, Jasinskas A, Obiero JM, Adenaiye O, Tai S, Hong F, Milton DK, Davies H, Felgner PL. Analysis of Serologic Cross-Reactivity Between Common Human Coronaviruses and SARS-CoV-2 Using Coronavirus Antigen Microarray. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.03.24.006544. [PMID: 32511324 PMCID: PMC7239054 DOI: 10.1101/2020.03.24.006544] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The current practice for diagnosis of SARS-CoV-2 infection relies on PCR testing of nasopharyngeal or respiratory specimens in a symptomatic patient at high epidemiologic risk. This testing strategy likely underestimates the true prevalence of infection, creating the need for serologic methods to detect infections missed by the limited testing to date. Here, we describe the development of a coronavirus antigen microarray containing immunologically significant antigens from SARS-CoV-2, in addition to SARS-CoV, MERS-CoV, common human coronavirus strains, and other common respiratory viruses. A preliminary study of human sera collected prior to the SARS-CoV-2 pandemic demonstrates overall high IgG reactivity to common human coronaviruses and low IgG reactivity to epidemic coronaviruses including SARS-CoV-2, with some cross-reactivity of conserved antigenic domains including S2 domain of spike protein and nucleocapsid protein. This array can be used to answer outstanding questions regarding SARS-CoV-2 infection, including whether baseline serology for other coronaviruses impacts disease course, how the antibody response to infection develops over time, and what antigens would be optimal for vaccine development.
Collapse
Affiliation(s)
- Saahir Khan
- Division of Infectious Diseases, Department of Medicine, University of California Irvine Health, Orange, CA
| | - Rie Nakajima
- Department of Physiology and Biophysics, School of Medicine, University of California Irvine, Irvine, CA
| | - Aarti Jain
- Department of Physiology and Biophysics, School of Medicine, University of California Irvine, Irvine, CA
| | - Rafael Ramiro de Assis
- Department of Physiology and Biophysics, School of Medicine, University of California Irvine, Irvine, CA
| | - Al Jasinskas
- Department of Physiology and Biophysics, School of Medicine, University of California Irvine, Irvine, CA
| | - Joshua M. Obiero
- Department of Physiology and Biophysics, School of Medicine, University of California Irvine, Irvine, CA
| | - Oluwasanmi Adenaiye
- Institute for Applied Environmental Health, School of Public Health, University of Maryland, College Park, MD
| | - Sheldon Tai
- Institute for Applied Environmental Health, School of Public Health, University of Maryland, College Park, MD
| | - Filbert Hong
- Institute for Applied Environmental Health, School of Public Health, University of Maryland, College Park, MD
| | - Donald K. Milton
- Institute for Applied Environmental Health, School of Public Health, University of Maryland, College Park, MD
| | - Huw Davies
- Department of Physiology and Biophysics, School of Medicine, University of California Irvine, Irvine, CA
| | - Philip L. Felgner
- Department of Physiology and Biophysics, School of Medicine, University of California Irvine, Irvine, CA
| | | |
Collapse
|
50
|
Menachery VD, Dinnon KH, Yount BL, McAnarney ET, Gralinski LE, Hale A, Graham RL, Scobey T, Anthony SJ, Wang L, Graham B, Randell SH, Lipkin WI, Baric RS. Trypsin Treatment Unlocks Barrier for Zoonotic Bat Coronavirus Infection. J Virol 2020; 94:e01774-19. [PMID: 31801868 PMCID: PMC7022341 DOI: 10.1128/jvi.01774-19] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/27/2019] [Indexed: 12/27/2022] Open
Abstract
Traditionally, the emergence of coronaviruses (CoVs) has been attributed to a gain in receptor binding in a new host. Our previous work with severe acute respiratory syndrome (SARS)-like viruses argued that bats already harbor CoVs with the ability to infect humans without adaptation. These results suggested that additional barriers limit the emergence of zoonotic CoV. In this work, we describe overcoming host restriction of two Middle East respiratory syndrome (MERS)-like bat CoVs using exogenous protease treatment. We found that the spike protein of PDF2180-CoV, a MERS-like virus found in a Ugandan bat, could mediate infection of Vero and human cells in the presence of exogenous trypsin. We subsequently show that the bat virus spike can mediate the infection of human gut cells but is unable to infect human lung cells. Using receptor-blocking antibodies, we show that infection with the PDF2180 spike does not require MERS-CoV receptor DPP4 and antibodies developed against the MERS spike receptor-binding domain and S2 portion are ineffective in neutralizing the PDF2180 chimera. Finally, we found that the addition of exogenous trypsin also rescues HKU5-CoV, a second bat group 2c CoV. Together, these results indicate that proteolytic cleavage of the spike, not receptor binding, is the primary infection barrier for these two group 2c CoVs. Coupled with receptor binding, proteolytic activation offers a new parameter to evaluate the emergence potential of bat CoVs and offers a means to recover previously unrecoverable zoonotic CoV strains.IMPORTANCE Overall, our studies demonstrate that proteolytic cleavage is the primary barrier to infection for a subset of zoonotic coronaviruses. Moving forward, the results argue that both receptor binding and proteolytic cleavage of the spike are critical factors that must be considered for evaluating the emergence potential and risk posed by zoonotic coronaviruses. In addition, the findings also offer a novel means to recover previously uncultivable zoonotic coronavirus strains and argue that other tissues, including the digestive tract, could be a site for future coronavirus emergence events in humans.
Collapse
Affiliation(s)
- Vineet D Menachery
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kenneth H Dinnon
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Boyd L Yount
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Eileen T McAnarney
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lisa E Gralinski
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Andrew Hale
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Rachel L Graham
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Trevor Scobey
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Simon J Anthony
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, New York, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | - Lingshu Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Barney Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Scott H Randell
- Department of Cell Biology and Physiology, and Marsico Lung Institute/Cystic Fibrosis Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - W Ian Lipkin
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, New York, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | - Ralph S Baric
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|