1
|
Roessler J, Pich D, Krähling V, Becker S, Keppler OT, Zeidler R, Hammerschmidt W. SARS-CoV-2 and Epstein-Barr Virus-like Particles Associate and Fuse with Extracellular Vesicles in Virus Neutralization Tests. Biomedicines 2023; 11:2892. [PMID: 38001893 PMCID: PMC10669694 DOI: 10.3390/biomedicines11112892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
The successful development of effective viral vaccines depends on well-known correlates of protection, high immunogenicity, acceptable safety criteria, low reactogenicity, and well-designed immune monitoring and serology. Virus-neutralizing antibodies are often a good correlate of protective immunity, and their serum concentration is a key parameter during the pre-clinical and clinical testing of vaccine candidates. Viruses are inherently infectious and potentially harmful, but we and others developed replication-defective SARS-CoV-2 virus-like-particles (VLPs) as surrogates for infection to quantitate neutralizing antibodies with appropriate target cells using a split enzyme-based approach. Here, we show that SARS-CoV-2 and Epstein-Barr virus (EBV)-derived VLPs associate and fuse with extracellular vesicles in a highly specific manner, mediated by the respective viral fusion proteins and their corresponding host receptors. We highlight the capacity of virus-neutralizing antibodies to interfere with this interaction and demonstrate a potent application using this technology. To overcome the common limitations of most virus neutralization tests, we developed a quick in vitro diagnostic assay based on the fusion of SARS-CoV-2 VLPs with susceptible vesicles to quantitate neutralizing antibodies without the need for infectious viruses or living cells. We validated this method by testing a set of COVID-19 patient serum samples, correlated the results with those of a conventional test, and found good sensitivity and specificity. Furthermore, we demonstrate that this serological assay can be adapted to a human herpesvirus, EBV, and possibly other enveloped viruses.
Collapse
Affiliation(s)
- Johannes Roessler
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany; (J.R.); (R.Z.)
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany;
- German Centre for Infection Research (DZIF), Partner Site Munich, 81377 Munich, Germany;
| | - Dagmar Pich
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany;
- German Centre for Infection Research (DZIF), Partner Site Munich, 81377 Munich, Germany;
| | - Verena Krähling
- Institute of Virology, Faculty of Medicine, Philipps University Marburg, 35043 Marburg, Germany; (V.K.); (S.B.)
- German Centre for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Stephan Becker
- Institute of Virology, Faculty of Medicine, Philipps University Marburg, 35043 Marburg, Germany; (V.K.); (S.B.)
- German Centre for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Oliver T. Keppler
- German Centre for Infection Research (DZIF), Partner Site Munich, 81377 Munich, Germany;
- COVID-19 Registry of the LMU Munich (CORKUM), LMU University Hospital, 81377 Munich, Germany
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Reinhard Zeidler
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany; (J.R.); (R.Z.)
- German Centre for Infection Research (DZIF), Partner Site Munich, 81377 Munich, Germany;
- Institute of Structural Biology, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Wolfgang Hammerschmidt
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany;
- German Centre for Infection Research (DZIF), Partner Site Munich, 81377 Munich, Germany;
| |
Collapse
|
2
|
Buffington J, Duan Z, Kwon HJ, Hong J, Li D, Feng M, Xie H, Ho M. Identification of nurse shark V NAR single-domain antibodies targeting the spike S2 subunit of SARS-CoV-2. FASEB J 2023; 37:e22973. [PMID: 37191949 PMCID: PMC10715488 DOI: 10.1096/fj.202202099rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 04/17/2023] [Accepted: 05/01/2023] [Indexed: 05/17/2023]
Abstract
SARS-CoV-2 is the etiological agent of the COVID-19 pandemic. Antibody-based therapeutics targeting the spike protein, specifically the S1 subunit or the receptor binding domain (RBD) of SARS-CoV-2, have gained attention due to their clinical efficacy in treating patients diagnosed with COVID-19. An alternative to conventional antibody therapeutics is the use of shark new antigen variable receptor domain (VNAR ) antibodies. VNAR s are small (<15 kDa) and can reach deep into the pockets or grooves of the target antigen. Here, we have isolated 53 VNAR s that bind to the S2 subunit by phage panning from a naïve nurse shark VNAR phage display library constructed in our laboratory. Among those binders, S2A9 showed the best neutralization activity against the original pseudotyped SARS-CoV-2 virus. Several binders, including S2A9, showed cross-reactivity against S2 subunits from other β coronaviruses. Furthermore, S2A9 showed neutralization activity against all variants of concern (VOCs) from alpha to omicron (including BA1, BA2, BA4, and BA5) in both pseudovirus and live virus neutralization assays. Our findings suggest that S2A9 could be a promising lead molecule for the development of broadly neutralizing antibodies against SARS-CoV-2 and emerging variants. The nurse shark VNAR phage library offers a novel platform that can be used to rapidly isolate single-domain antibodies against emerging viral pathogens.
Collapse
Affiliation(s)
- Jesse Buffington
- Antibody Engineering Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Zhijian Duan
- Antibody Engineering Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hyung Joon Kwon
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jessica Hong
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Dan Li
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mingqian Feng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hang Xie
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Mitchell Ho
- Antibody Engineering Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Yonker LM, Swank Z, Bartsch YC, Burns MD, Kane A, Boribong BP, Davis JP, Loiselle M, Novak T, Senussi Y, Cheng CA, Burgess E, Edlow AG, Chou J, Dionne A, Balaguru D, Lahoud-Rahme M, Arditi M, Julg B, Randolph AG, Alter G, Fasano A, Walt DR. Circulating Spike Protein Detected in Post-COVID-19 mRNA Vaccine Myocarditis. Circulation 2023; 147:867-876. [PMID: 36597886 PMCID: PMC10010667 DOI: 10.1161/circulationaha.122.061025] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 11/23/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Cases of adolescents and young adults developing myocarditis after vaccination with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-targeted mRNA vaccines have been reported globally, but the underlying immunoprofiles of these individuals have not been described in detail. METHODS From January 2021 through February 2022, we prospectively collected blood from 16 patients who were hospitalized at Massachusetts General for Children or Boston Children's Hospital for myocarditis, presenting with chest pain with elevated cardiac troponin T after SARS-CoV-2 vaccination. We performed extensive antibody profiling, including tests for SARS-CoV-2-specific humoral responses and assessment for autoantibodies or antibodies against the human-relevant virome, SARS-CoV-2-specific T-cell analysis, and cytokine and SARS-CoV-2 antigen profiling. Results were compared with those from 45 healthy, asymptomatic, age-matched vaccinated control subjects. RESULTS Extensive antibody profiling and T-cell responses in the individuals who developed postvaccine myocarditis were essentially indistinguishable from those of vaccinated control subjects, despite a modest increase in cytokine production. A notable finding was that markedly elevated levels of full-length spike protein (33.9±22.4 pg/mL), unbound by antibodies, were detected in the plasma of individuals with postvaccine myocarditis, whereas no free spike was detected in asymptomatic vaccinated control subjects (unpaired t test; P<0.0001). CONCLUSIONS Immunoprofiling of vaccinated adolescents and young adults revealed that the mRNA vaccine-induced immune responses did not differ between individuals who developed myocarditis and individuals who did not. However, free spike antigen was detected in the blood of adolescents and young adults who developed post-mRNA vaccine myocarditis, advancing insight into its potential underlying cause.
Collapse
Affiliation(s)
- Lael M. Yonker
- Mucosal Immunology and Biology Research Center (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Department of Pediatrics (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., D.B., M.L.-R., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
| | - Zoe Swank
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA (Z.S., Y.S., C.-A.C., D.R.W.)
| | - Yannic C. Bartsch
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA (Y.C.B., E.B., B.J., G.A.)
| | - Madeleine D. Burns
- Mucosal Immunology and Biology Research Center (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Department of Pediatrics (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., D.B., M.L.-R., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
| | - Abigail Kane
- Mucosal Immunology and Biology Research Center (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Department of Pediatrics (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., D.B., M.L.-R., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
| | - Brittany P. Boribong
- Mucosal Immunology and Biology Research Center (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Department of Pediatrics (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., D.B., M.L.-R., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
| | - Jameson P. Davis
- Mucosal Immunology and Biology Research Center (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Department of Pediatrics (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., D.B., M.L.-R., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
| | - Maggie Loiselle
- Mucosal Immunology and Biology Research Center (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Department of Pediatrics (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., D.B., M.L.-R., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
| | - Tanya Novak
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
- Department of Anesthesiology, Critical Care and Pain Medicine (T.N., A.G.R.), Boston Children’s Hospital, MA
| | - Yasmeen Senussi
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA (Z.S., Y.S., C.-A.C., D.R.W.)
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA (Z.S., Y.S., C.-A.C., D.R.W.)
| | - Chi-An Cheng
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA (Z.S., Y.S., C.-A.C., D.R.W.)
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA (Z.S., Y.S., C.-A.C., D.R.W.)
| | - Eleanor Burgess
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA (Y.C.B., E.B., B.J., G.A.)
| | - Andrea G. Edlow
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology (A.G.E.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Vincent Center for Reproductive Biology (A.G.E.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Department of Anesthesiology, Critical Care and Pain Medicine (T.N., A.G.R.), Boston Children’s Hospital, MA
| | - Janet Chou
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
- Department of Pediatrics, Division of Immunology (J.C.), Boston Children’s Hospital, MA
| | - Audrey Dionne
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
- Department of Cardiology (A.D.), Boston Children’s Hospital, MA
| | - Duraisamy Balaguru
- Department of Pediatrics (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., D.B., M.L.-R., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
| | - Manuella Lahoud-Rahme
- Department of Pediatrics (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., D.B., M.L.-R., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Infectious and Immunologic Diseases Research Center, and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA (M.A.)
| | - Boris Julg
- Department of Medicine (B.J.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA (Y.C.B., E.B., B.J., G.A.)
| | - Adrienne G. Randolph
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
| | - Galit Alter
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA (Y.C.B., E.B., B.J., G.A.)
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Department of Pediatrics (L.M.Y., M.D.B., A.K., B.P.B., J.P.D., M.L., D.B., M.L.-R., A.F.), Division of Infectious Disease, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
| | - David R. Walt
- Harvard Medical School, Boston, MA (L.M.Y., Z.S., Y.C.B., B.P.B., T.N., Y.S., C.-A.C., J.C., A.D., D.B., M.L.-R., B.J., A.G.R., G.A., A.F., D.R.W.)
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA (Z.S., Y.S., C.-A.C., D.R.W.)
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA (Z.S., Y.S., C.-A.C., D.R.W.)
| |
Collapse
|
4
|
Ng KW, Faulkner N, Finsterbusch K, Wu M, Harvey R, Hussain S, Greco M, Liu Y, Kjaer S, Swanton C, Gandhi S, Beale R, Gamblin SJ, Cherepanov P, McCauley J, Daniels R, Howell M, Arase H, Wack A, Bauer DLV, Kassiotis G. SARS-CoV-2 S2-targeted vaccination elicits broadly neutralizing antibodies. Sci Transl Med 2022; 14:eabn3715. [PMID: 35895836 DOI: 10.1126/scitranslmed.abn3715] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Several variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have emerged during the current coronavirus disease 2019 (COVID-19) pandemic. Although antibody cross-reactivity with the spike glycoproteins (S) of diverse coronaviruses, including endemic common cold coronaviruses (HCoVs), has been documented, it remains unclear whether such antibody responses, typically targeting the conserved S2 subunit, contribute to protection when induced by infection or through vaccination. Using a mouse model, we found that prior HCoV-OC43 S-targeted immunity primes neutralizing antibody responses to otherwise subimmunogenic SARS-CoV-2 S exposure and promotes S2-targeting antibody responses. Moreover, vaccination with SARS-CoV-2 S2 elicited antibodies in mice that neutralized diverse animal and human alphacoronaviruses and betacoronaviruses in vitro and provided a degree of protection against SARS-CoV-2 challenge in vivo. Last, in mice with a history of SARS-CoV-2 Wuhan-based S vaccination, further S2 vaccination induced broader neutralizing antibody response than booster Wuhan S vaccination, suggesting that it may prevent repertoire focusing caused by repeated homologous vaccination. These data establish the protective value of an S2-targeting vaccine and support the notion that S2 vaccination may better prepare the immune system to respond to the changing nature of the S1 subunit in SARS-CoV-2 variants of concern, as well as to future coronavirus zoonoses.
Collapse
Affiliation(s)
- Kevin W Ng
- Retroviral Immunology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Nikhil Faulkner
- Retroviral Immunology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK
| | - Katja Finsterbusch
- Immunoregulation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Mary Wu
- High Throughput Screening STP, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ruth Harvey
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Saira Hussain
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- RNA Virus Replication Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Maria Greco
- RNA Virus Replication Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Yafei Liu
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Laboratory of Immunochemistry, World Premier International Immunology Frontier Research Centre, Osaka University, Osaka 565-0871, Japan
| | - Svend Kjaer
- Structural Biology STP, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
| | - Sonia Gandhi
- Neurodegradation Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Rupert Beale
- Cell Biology of Infection Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Steve J Gamblin
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Peter Cherepanov
- Chromatin structure and mobile DNA Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - John McCauley
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Rodney Daniels
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Michael Howell
- High Throughput Screening STP, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Hisashi Arase
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Laboratory of Immunochemistry, World Premier International Immunology Frontier Research Centre, Osaka University, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
| | - Andreas Wack
- Immunoregulation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - David L V Bauer
- RNA Virus Replication Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - George Kassiotis
- Retroviral Immunology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Infectious Disease, St Mary's Hospital, Imperial College London, London W2 1PG, UK
| |
Collapse
|
5
|
IgG targeting distinct seasonal coronavirus- conserved SARS-CoV-2 spike subdomains correlates with differential COVID-19 disease outcomes. Cell Rep 2022; 39:110904. [PMID: 35617962 PMCID: PMC9108089 DOI: 10.1016/j.celrep.2022.110904] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/25/2022] [Accepted: 05/11/2022] [Indexed: 11/20/2022] Open
Abstract
Despite SARS-CoV-2 being a "novel" virus, early detection of anti-spike IgG in severe COVID-19 patients may be caused by the amplification of humoral memory responses against seasonal coronaviruses. Here, we examine this phenomenon by characterizing anti-spike IgG responses in non-hospitalized convalescent individuals across a spectrum of COVID-19 severity. We observe that disease severity positively correlates with anti-spike IgG levels, IgG cross-reactivity against other betacoronaviruses (β-CoVs), and FcγR activation. Analysis of IgG targeting β-CoV-conserved and non-conserved immunodominant epitopes within the SARS-CoV-2 spike protein revealed epitope-specific relationships: IgG targeting the conserved heptad repeat (HR) 2 region significantly correlates with milder disease, while targeting the conserved S2'FP region correlates with more severe disease. Furthermore, a lower HR2-to-S2'FP IgG-binding ratio correlates with greater disease severity, with ICU-hospitalized COVID-19 patients showing the lowest HR2/S2'FP ratios. These findings suggest that HR2/S2'FP IgG profiles may predict disease severity and offer insight into protective versus deleterious humoral recall responses.
Collapse
|
6
|
Sepand MR, Bigdelou B, Ho JQ, Sharaf M, Lannigan AJ, Sullivan IM, da Silva AP, Barrett LO, McGoldrick S, Lnu Y, Lynch SE, Boisclair JM, Barnard-Pratt DD, Zanganeh S. Long-Term Immunity and Antibody Response: Challenges for Developing Efficient COVID-19 Vaccines. Antibodies (Basel) 2022; 11:35. [PMID: 35645208 PMCID: PMC9149948 DOI: 10.3390/antib11020035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/01/2022] [Accepted: 05/06/2022] [Indexed: 12/04/2022] Open
Abstract
Questions and concerns regarding the efficacy and immunogenicity of coronavirus disease 2019 (COVID-19) vaccines have plagued scientists since the BNT162b2 mRNA vaccine was introduced in late 2020. As a result, decisions about vaccine boosters based on breakthrough infection rates and the decline of antibody titers have commanded worldwide attention and research. COVID-19 patients have displayed continued severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-spike-protein-specific antibodies and neutralizing antibodies in longitudinal studies; in addition, cytokine activation has been detected at early steps following SARS-CoV-2 infection. Epitopes that are highly reactive and can mediate long-term antibody responses have been identified at the spike and ORF1ab proteins. The N-terminal domain of the S1 and S2 subunits is the location of important SARS-CoV-2 spike protein epitopes. High sequence identity between earlier and newer variants of SARS-CoV-2 and different degrees of sequence homology among endemic human coronaviruses have been observed. Understanding the extent and duration of protective immunity is consequential for determining the course of the COVID-19 pandemic. Further knowledge of memory responses to different variants of SARS-CoV-2 is needed to improve the design of the vaccine.
Collapse
Affiliation(s)
- Mohammad Reza Sepand
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Banafsheh Bigdelou
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Jim Q. Ho
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Mohammad Sharaf
- Department of Chemical and Biomolecular Engineering, New York University, New York, NY 10012, USA;
| | - Alexis J. Lannigan
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Ian M. Sullivan
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Alecsander P. da Silva
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Leland O. Barrett
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Scott McGoldrick
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Yuvraj Lnu
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Shannon E. Lynch
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Jared M. Boisclair
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Dakarai D. Barnard-Pratt
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Steven Zanganeh
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| |
Collapse
|
7
|
Roessler J, Pich D, Albanese M, Wratil PR, Krähling V, Hellmuth JC, Scherer C, von Bergwelt-Baildon M, Becker S, Keppler OT, Brisson A, Zeidler R, Hammerschmidt W. Quantitation of SARS-CoV-2 neutralizing antibodies with a virus-free, authentic test. PNAS NEXUS 2022; 1:pgac045. [PMID: 36382127 PMCID: PMC9645495 DOI: 10.1093/pnasnexus/pgac045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/02/2022] [Accepted: 04/11/2022] [Indexed: 06/16/2023]
Abstract
Neutralizing antibodies (NAbs), and their concentration in sera of convalescents and vaccinees are a correlate of protection from COVID-19. The antibody concentrations in clinical samples that neutralize SARS-CoV-2 are difficult and very cumbersome to assess with conventional virus neutralization tests (cVNTs), which require work with the infectious virus and biosafety level 3 containment precautions. Alternative virus neutralization tests currently in use are mostly surrogate tests based on direct or competitive enzyme immunoassays or use viral vectors with the spike protein as the single structural component of SARS-CoV-2. To overcome these obstacles, we developed a virus-free, safe and very fast (4.5 h) in vitro diagnostic test based on engineered yet authentic SARS-CoV-2 virus-like-particles (VLPs). They share all features of the original SARS-CoV-2 but lack the viral RNA genome and thus are non-infectious. NAbs induced by infection or vaccination, but also potentially neutralizing monoclonal antibodies can be reliably quantified and assessed with ease and within hours with our test, because they interfere and block the ACE2-mediated uptake of VLPs by recipient cells. Results from the VLP neutralization test (VLPNT) showed excellent specificity and sensitivity and correlated very well with a cVNT using fully infectious SARS-CoV-2. The results also demonstrated the reduced neutralizing capacity of COVID-19 vaccinee sera against variants of concern of SARS-CoV-2 including omicron B.1.1.529, BA.1.
Collapse
Affiliation(s)
- Johannes Roessler
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- German Centre for Infection Research (DZIF), Partner site Munich, Germany
| | - Dagmar Pich
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- German Centre for Infection Research (DZIF), Partner site Munich, Germany
| | - Manuel Albanese
- German Centre for Infection Research (DZIF), Partner site Munich, Germany
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Paul R Wratil
- German Centre for Infection Research (DZIF), Partner site Munich, Germany
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Verena Krähling
- Institute of Virology, Faculty of Medicine, Philipps-Universität Marburg, Marburg, Germany
- German Centre for Infection Research (DZIF), Partner site Giessen-Marburg-Langen, Marburg, Germany
| | - Johannes C Hellmuth
- Department of Medicine III, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Clemens Scherer
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Medicine I, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- German Cancer Consortium (DKTK), Munich, Germany
| | - Stephan Becker
- Institute of Virology, Faculty of Medicine, Philipps-Universität Marburg, Marburg, Germany
- German Centre for Infection Research (DZIF), Partner site Giessen-Marburg-Langen, Marburg, Germany
| | - Oliver T Keppler
- German Centre for Infection Research (DZIF), Partner site Munich, Germany
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Alain Brisson
- UMR-CBMN CNRS-University of Bordeaux-INP, Pessac, France
| | - Reinhard Zeidler
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- German Centre for Infection Research (DZIF), Partner site Munich, Germany
| | - Wolfgang Hammerschmidt
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
- German Centre for Infection Research (DZIF), Partner site Munich, Germany
| |
Collapse
|
8
|
Lerdsamran H, Mungaomklang A, Iamsirithaworn S, Prasertsopon J, Wiriyarat W, Saritsiri S, Anusorntanawat R, Siriyakorn N, Intalapaporn P, Sirikhetkon S, Sangsiriwut K, Dangsakul W, Sawadpongpan S, Thinpan N, Kitidee K, Okada P, Techasuwanna R, Mongkalangoon N, Prasert K, Puthavathana P. Seroprevalence of anti-SARS-CoV-2 antibodies in Thai adults during the first three epidemic waves. PLoS One 2022; 17:e0263316. [PMID: 35476709 PMCID: PMC9045619 DOI: 10.1371/journal.pone.0263316] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/28/2022] [Indexed: 12/23/2022] Open
Abstract
This study determined the presence of anti-SARS-CoV-2 antibodies in 4964 individuals, comprising 300 coronavirus disease-19 (COVID-19) prepandemic serum samples, 142 COVID-19 patients, 2113 individuals at risk due to their occupations, 1856 individuals at risk due to sharing workplaces or communities with COVID-19 patients, and 553 Thai citizens returning after spending extended periods of time in countries with a high disease prevalence. We recruited participants between May 2020 and May 2021, which spanned the first two epidemic waves and part of the third wave of the COVID-19 outbreaks in Thailand. Their sera were tested in a microneutralization and a chemiluminescence immunoassay for IgG against the N protein. Furthermore, we performed an immunofluorescence assay to resolve discordant results between the two assays. None of the prepandemic sera contained anti-SARS-CoV-2 antibodies, while antibodies developed in 88% (15 of 17) of the COVID-19 patients at 8–14 days and in 94–100% of the patients between 15 and 60 days after disease onset. Neutralizing antibodies persisted for at least 8 months, longer than IgG antibodies. Of the 2113 individuals at risk due to their occupation, none of the health providers, airport officers, or public transport drivers were seropositive, while antibodies were present in 0.44% of entertainment workers. Among the 1856 individuals at risk due to sharing workplaces or communities with COVID-19 patients, seropositivity was present in 1.9, 1.5, and 7.5% of the Bangkok residents during the three epidemic waves, respectively, and in 1.3% of the Chiang Mai people during the first epidemic wave. The antibody prevalence varied between 6.5 and 47.0% in 553 Thai people returning from high-risk countries. This serosurveillance study found a low infection rate of SARS-CoV-2 in Thailand before the emergence of the Delta variant in late May 2021. The findings support the Ministry of Public Health’s data, which are based on numbers of patients and contact tracing.
Collapse
Affiliation(s)
- Hatairat Lerdsamran
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Anek Mungaomklang
- Institute for Urban Disease Control and Prevention, Department of Disease Control, Ministry of Public Health, Bangkok, Thailand
| | | | - Jarunee Prasertsopon
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | | | - Suthee Saritsiri
- The 67th Public Health Center Thaweewatthana, Department of Health, Bangkok Metropolitan Administration, Bangkok, Thailand
| | - Ratikorn Anusorntanawat
- Chaophraya Yommarat Hospital, Office of the Permanent Secretary, Ministry of Public Health, Suphanburi, Thailand
| | - Nirada Siriyakorn
- Rajavithi Hospital, Department of Medical Services, Ministry of Public Health, Bangkok, Thailand
| | - Poj Intalapaporn
- Rajavithi Hospital, Department of Medical Services, Ministry of Public Health, Bangkok, Thailand
| | - Somrak Sirikhetkon
- Institute for Urban Disease Control and Prevention, Department of Disease Control, Ministry of Public Health, Bangkok, Thailand
| | - Kantima Sangsiriwut
- Department of Preventive and Social Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Worawat Dangsakul
- Department of Medical Science, Ministry of Public Health, Nonthaburi, Thailand
| | - Suteema Sawadpongpan
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Nattakan Thinpan
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Kuntida Kitidee
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Pilailuk Okada
- Department of Medical Science, Ministry of Public Health, Nonthaburi, Thailand
| | - Ranida Techasuwanna
- Department of Disease Control, Ministry of Public Health, Nonthaburi, Thailand
| | | | - Kriengkrai Prasert
- Nakhon Phanom Provincial Hospital, Department of Medical Services, Ministry of Public Health, Nakhon Phanom, Thailand
| | - Pilaipan Puthavathana
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
- * E-mail:
| |
Collapse
|
9
|
Xiang R, Wang Y, Wang L, Deng X, Huo S, Jiang S, Yu F. Neutralizing monoclonal antibodies against highly pathogenic coronaviruses. Curr Opin Virol 2022; 53:101199. [PMID: 35038651 PMCID: PMC8716168 DOI: 10.1016/j.coviro.2021.12.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 12/17/2021] [Accepted: 12/24/2021] [Indexed: 12/15/2022]
Abstract
The pandemic of Coronavirus Disease 2019 (COVID-19) caused by severe acute respiratory syndrome 2 coronavirus (SARS-CoV-2) is a continuing worldwide threat to human health and social economy. Historically, SARS-CoV-2 follows SARS and MERS as the third coronavirus spreading across borders and continents, but far more dangerous with long-lasting symptomatic consequences. The current situation is strong evidence that coronaviruses will continue to be pathogens of consequence in the future, thus calling for the development of neutralizing antibody-based prophylactics and therapeutics for prevention and treatment of COVID-19 and other human coronavirus diseases. This review summarized the progresses of developing neutralizing monoclonal antibodies against infection of SARS-CoV-2, SARS-CoV, and MERS-CoV, and discussed their potential applications in prevention and treatment of COVID-19 and other human coronavirus diseases.
Collapse
Affiliation(s)
- Rong Xiang
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Yang Wang
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Lili Wang
- Research Center of Chinese Jujube, Hebei Agricultural University, Baoding, China
| | - Xiaoqian Deng
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Shanshan Huo
- College of Life Sciences, Hebei Agricultural University, Baoding, China; Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Baoding, China.
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
| | - Fei Yu
- College of Life Sciences, Hebei Agricultural University, Baoding, China; Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Baoding, China.
| |
Collapse
|
10
|
Abstract
Most of SARS-CoV-2 neutralizing antibodies (nAbs) targeted the receptor binding domain (RBD) of the SARS-CoV-2 spike (S) protein. However, mutations at RBD sequences found in the emerging SARS-CoV-2 variants greatly reduced the effectiveness of nAbs. Here we showed that four nAbs, S2-4D, S2-5D, S2-8D, and S2-4A, which recognized a conserved epitope in the S2 subunit of the S protein, can inhibit SARS-CoV-2 infection through blocking the S protein-mediated membrane fusion. Notably, these four nAbs exhibited broadly neutralizing activity against SARS-CoV-2 Alpha, Gamma, Delta, and Epsilon variants. Antisera collected from mice immunized with the identified epitope peptides of these four nAbs also exhibited potent virus neutralizing activity. Discovery of the S2-specific nAbs and their unique antigenic epitopes paves a new path for development of COVID-19 therapeutics and vaccines. IMPORTANCE The spike (S) protein on the surface of SARS-CoV-2 mediates receptor binding and virus-host cell membrane fusion during virus entry. Many neutralizing antibodies (nAbs), which targeted the receptor binding domain (RBD) of S protein, lost the neutralizing activity against the newly emerging SARS-CoV-2 variants with sequence mutations at the RBD. In contrast, the nAb against the highly conserved S2 subunit, which plays the key role in virus–host cell membrane fusion, was poorly discovered. We showed that four S2-specific nAbs, S2-4D, S2-5D, S2-8D, and S2-4A, inhibited SARS-CoV-2 infection through blocking the S protein-mediated membrane fusion. These nAbs exhibited broadly neutralizing activity against Alpha, Gamma, Delta, and Epsilon variants. Antisera induced by the identified epitope peptides also possessed potent neutralizing activity. This work not only unveiled the S2-specific nAbs but also discovered an immunodominant epitope in the S2 subunit that can be rationally designed as the broad-spectrum vaccine against the SARS-like coronaviruses.
Collapse
|
11
|
Lerdsamran H, Mungaomklang A, Iamsirithaworn S, Prasertsopon J, Prasert K, Intalapaporn P, Siriyakorn N, Wiriyarat W, Thinpan N, Sawadpongpan S, Sirikhetkon S, Mongkalangoon N, Petto S, Puthavathana P. Evaluation of different platforms for the detection of anti-SARS coronavirus-2 antibodies, Thailand. BMC Infect Dis 2021; 21:1213. [PMID: 34872510 PMCID: PMC8646009 DOI: 10.1186/s12879-021-06921-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 11/29/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) help determine previous infection in individuals, regardless of whether they are asymptomatic or symptomatic. The detection of antibodies serves several purposes, including supporting other assays for disease diagnosis, conducting seroepidemiological studies, and evaluating vaccines. Many platforms of immunological methods for anti-SARS-CoV-2 antibody detection and their performance require validation. METHODS This study evaluated the test performance of three autoanalyzer-based assays (Architect IgG, Vitros IgG, and Vitros total Ig) and one manual ELISA (Wantai total Ig) against a microneutralization (microNT) assay on the detection of SARS-CoV-2 antibodies. Furthermore, an indirect immunofluorescence assay verified the discordant results between the microNT and commercial assays. The test sensitivity, specificity, positive predictive value, and negative predictive value were determined based on four groups of 1005 serum samples: 102 COVID-19 prepandemic sera, 45 anti-SARS-CoV-2 positive sera, 366 sera of people at risk, and 492 sera of citizens returning from countries with a high prevalence of infection. RESULTS The analyses as a whole showed that the performance of these commercial assays was comparable. Each group was also analysed separately to gain further insight into test performance. The Architect did not detect two positive sera of people at risk (prevalence of infection 0.55%). The other methods correctly identified these two positive sera but yielded varying false-positive results. The group of returning travellers with an infection rate of 28.3% (139 of 492) better differentiated the test performance of individual assays. CONCLUSIONS High-throughput Architect and Vitros autoanalyzers appear appropriate for working on large sample sizes in countries that can afford the cost. The Wantai ELISA, while requiring more individual time and technical skill, may provide reliable results at a lower cost. The selection of assays will depend on the laboratory facilities and feasibility.
Collapse
Affiliation(s)
- Hatairat Lerdsamran
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Anek Mungaomklang
- Institute for Urban Disease Control and Prevention, Department of Disease Control, Ministry of Public Health, Bangkok, 10220, Thailand
| | - Sopon Iamsirithaworn
- Department of Disease Control, Ministry of Public Health, Nonthaburi, 11000, Thailand
| | - Jarunee Prasertsopon
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Kriengkrai Prasert
- Department of Medical Services, Nakhon Phanom Provincial Hospital, Ministry of Public Health, Nakhon Phanom, 48000, Thailand
| | - Poj Intalapaporn
- Division of Infectious Disease, Department of Medicine, Rajavithi Hospital, Ministry of Public Health, Bangkok, 10400, Thailand
| | - Nirada Siriyakorn
- Division of Infectious Disease, Department of Medicine, Rajavithi Hospital, Ministry of Public Health, Bangkok, 10400, Thailand
| | - Witthawat Wiriyarat
- Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Nattakan Thinpan
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Suteema Sawadpongpan
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Somrak Sirikhetkon
- Institute for Urban Disease Control and Prevention, Department of Disease Control, Ministry of Public Health, Bangkok, 10220, Thailand
| | - Noparat Mongkalangoon
- Department of Disease Control, Ministry of Public Health, Nonthaburi, 11000, Thailand
| | - Suwanna Petto
- Institute for Urban Disease Control and Prevention, Department of Disease Control, Ministry of Public Health, Bangkok, 10220, Thailand
| | - Pilaipan Puthavathana
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
12
|
Ng KW, Faulkner N, Wrobel AG, Gamblin SJ, Kassiotis G. Heterologous humoral immunity to human and zoonotic coronaviruses: Aiming for the achilles heel. Semin Immunol 2021; 55:101507. [PMID: 34716096 PMCID: PMC8542444 DOI: 10.1016/j.smim.2021.101507] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 02/04/2023]
Abstract
Coronaviruses are evolutionarily successful RNA viruses, common to multiple avian, amphibian and mammalian hosts. Despite their ubiquity and potential impact, knowledge of host immunity to coronaviruses remains incomplete, partly owing to the lack of overt pathogenicity of endemic human coronaviruses (HCoVs), which typically cause common colds. However, the need for deeper understanding became pressing with the zoonotic introduction of three novel coronaviruses in the past two decades, causing severe acute respiratory syndromes in humans, and the unfolding pandemic of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This renewed interest not only triggered the discovery of two of the four HCoVs, but also uncovered substantial cellular and humoral cross-reactivity with shared or related coronaviral antigens. Here, we review the evidence for cross-reactive B cell memory elicited by HCoVs and its potential impact on the puzzlingly variable outcome of SARS-CoV-2 infection. The available data indicate targeting of highly conserved regions primarily in the S2 subunits of the spike glycoproteins of HCoVs and SARS-CoV-2 by cross-reactive B cells and antibodies. Rare monoclonal antibodies reactive with conserved S2 epitopes and with potent virus neutralising activity have been cloned, underscoring the potential functional relevance of cross-reactivity. We discuss B cell and antibody cross-reactivity in the broader context of heterologous humoral immunity to coronaviruses, as well as the limits of protective immune memory against homologous re-infection. Given the bidirectional nature of cross-reactivity, the unprecedented current vaccination campaign against SARS-CoV-2 is expected to impact HCoVs, as well as future zoonotic coronaviruses attempting to cross the species barrier. However, emerging SARS-CoV-2 variants with resistance to neutralisation by vaccine-induced antibodies highlight a need for targeting more constrained, less mutable parts of the spike. The delineation of such cross-reactive areas, which humoral immunity can be trained to attack, may offer the key to permanently shifting the balance of our interaction with current and future coronaviruses in our favour.
Collapse
Affiliation(s)
- Kevin W Ng
- Retroviral Immunology Laboratory, London, NW1 1AT, UK
| | - Nikhil Faulkner
- Retroviral Immunology Laboratory, London, NW1 1AT, UK; National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Antoni G Wrobel
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Steve J Gamblin
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - George Kassiotis
- Retroviral Immunology Laboratory, London, NW1 1AT, UK; Department of Infectious Disease, St Mary's Hospital, Imperial College London, London W2 1PG, UK.
| |
Collapse
|
13
|
El-Sayed A, Kamel M. Coronaviruses in humans and animals: the role of bats in viral evolution. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:19589-19600. [PMID: 33655480 PMCID: PMC7924989 DOI: 10.1007/s11356-021-12553-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/14/2021] [Indexed: 04/15/2023]
Abstract
Bats act as a natural reservoir for many viruses, including coronaviruses, and have played a crucial epidemiological role in the emergence of many viral diseases. Coronaviruses have been known for 60 years. They are usually responsible for the induction of mild respiratory signs in humans. However, since 2002, the bat-borne virus started to induce fatal epidemics according to WHO reports. In this year, the first serious human coronavirus epidemic (severe acute respiratory syndrome; SARS) occurred (China, 8098 cases, 774 deaths [9.5% of the cases] in 17 countries). The case fatality was higher in elderly patients above 60 years and reached 50% of the cases. SARS epidemic was followed 10 years later by the emergence of the middle east respiratory syndrome (MERS) in Saudi Arabia (in 2012, 2260 cases, 803 deaths [35.5% of the cases] in 27 countries). Finally, in December 2019, a new epidemic in Wuhan, China, (corona virus disease 2019, COVID-19) emerged and could spread to 217 countries infecting more than 86,255,226 cases and killing 1,863,973 people by the end of 2020. There are many reasons why bats are ideal reservoir hosts for viral diseases such as the tolerance of their immune system to the invading viruses for several months. They can actively shed the viruses, although they develop no clinical signs (will be discussed in details later in the review). Bats were directly or indirectly involved in the three previous coronavirus epidemics. The indirect transmission takes place via intermediate hosts including civet cats for SARS and dromedary camels in the case of MERS. Although bats are believed to be the source of COVID-19 pandemic, direct pieces of evidence are still lacking. Therefore, coronaviruses' role in epidemics induction and the epidemiological role of bats are discussed. The current work also presents different evidence (phylogenetic data, animal experiments, bats artificial infection studies, and computerized models of SARS-CoV2 evolution) that underline the involvement of bats in the epidemiology of the pandemic.
Collapse
Affiliation(s)
- Amr El-Sayed
- Department of Medicine and Infectious Diseases, Faculty of Medicine and Infectious Diseases, Cairo University, Giza, 12211, Egypt
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Medicine and Infectious Diseases, Cairo University, Giza, 12211, Egypt.
| |
Collapse
|
14
|
Villa TG, Abril AG, Sánchez S, de Miguel T, Sánchez-Pérez A. Animal and human RNA viruses: genetic variability and ability to overcome vaccines. Arch Microbiol 2021; 203:443-464. [PMID: 32989475 PMCID: PMC7521576 DOI: 10.1007/s00203-020-02040-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/29/2020] [Accepted: 09/12/2020] [Indexed: 02/06/2023]
Abstract
RNA viruses, in general, exhibit high mutation rates; this is mainly due to the low fidelity displayed by the RNA-dependent polymerases required for their replication that lack the proofreading machinery to correct misincorporated nucleotides and produce high mutation rates. This lack of replication fidelity, together with the fact that RNA viruses can undergo spontaneous mutations, results in genetic variants displaying different viral morphogenesis, as well as variation on their surface glycoproteins that affect viral antigenicity. This diverse viral population, routinely containing a variety of mutants, is known as a viral 'quasispecies'. The mutability of their virions allows for fast evolution of RNA viruses that develop antiviral resistance and overcome vaccines much more rapidly than DNA viruses. This also translates into the fact that pathogenic RNA viruses, that cause many diseases and deaths in humans, represent the major viral group involved in zoonotic disease transmission, and are responsible for worldwide pandemics.
Collapse
Affiliation(s)
- T G Villa
- Department of Microbiology, Faculty of Pharmacy, University of Santiago de Compostela, 5706, Santiago de Compostela, Spain.
| | - Ana G Abril
- Department of Microbiology, Faculty of Pharmacy, University of Santiago de Compostela, 5706, Santiago de Compostela, Spain
| | - S Sánchez
- Department of Microbiology, Faculty of Pharmacy, University of Santiago de Compostela, 5706, Santiago de Compostela, Spain
| | - T de Miguel
- Department of Microbiology, Faculty of Pharmacy, University of Santiago de Compostela, 5706, Santiago de Compostela, Spain
| | - A Sánchez-Pérez
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
15
|
Focosi D, Maggi F, Mazzetti P, Pistello M. Viral infection neutralization tests: A focus on severe acute respiratory syndrome-coronavirus-2 with implications for convalescent plasma therapy. Rev Med Virol 2021; 31:e2170. [PMID: 33350017 PMCID: PMC7536930 DOI: 10.1002/rmv.2170] [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: 08/12/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/15/2022]
Abstract
Viral neutralization tests (VNTs) have long been considered old-fashioned tricks in the armamentarium of fundamental virology, with laboratory implementation for a limited array of viruses only. Nevertheless, they represent the most reliable surrogate of potency for passive immunotherapies, such as monoclonal or polyclonal antibody therapy. The recent interest around therapy with convalescent plasma or monoclonal antibodies for the Covid-19 pandemic has paralleled the revival of VNTs. We review here the available methods by dissecting variations for each fundamental component of the VNT (i.e., virus type and dose, replication-competent cell line, serum, and detection system).
Collapse
Affiliation(s)
- Daniele Focosi
- North‐Western Tuscany Blood BankPisa University HospitalPisaItaly
| | | | | | - Mauro Pistello
- Department of Translational ResearchUniversity of PisaPisaItaly
| |
Collapse
|
16
|
Ricke DO. Two Different Antibody-Dependent Enhancement (ADE) Risks for SARS-CoV-2 Antibodies. Front Immunol 2021; 12:640093. [PMID: 33717193 PMCID: PMC7943455 DOI: 10.3389/fimmu.2021.640093] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/03/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 (SARS-CoV-2) disease severity and stages varies from asymptomatic, mild flu-like symptoms, moderate, severe, critical, and chronic disease. COVID-19 disease progression include lymphopenia, elevated proinflammatory cytokines and chemokines, accumulation of macrophages and neutrophils in lungs, immune dysregulation, cytokine storms, acute respiratory distress syndrome (ARDS), etc. Development of vaccines to severe acute respiratory syndrome (SARS), Middle East Respiratory Syndrome coronavirus (MERS-CoV), and other coronavirus has been difficult to create due to vaccine induced enhanced disease responses in animal models. Multiple betacoronaviruses including SARS-CoV-2 and SARS-CoV-1 expand cellular tropism by infecting some phagocytic cells (immature macrophages and dendritic cells) via antibody bound Fc receptor uptake of virus. Antibody-dependent enhancement (ADE) may be involved in the clinical observation of increased severity of symptoms associated with early high levels of SARS-CoV-2 antibodies in patients. Infants with multisystem inflammatory syndrome in children (MIS-C) associated with COVID-19 may also have ADE caused by maternally acquired SARS-CoV-2 antibodies bound to mast cells. ADE risks associated with SARS-CoV-2 has implications for COVID-19 and MIS-C treatments, B-cell vaccines, SARS-CoV-2 antibody therapy, and convalescent plasma therapy for patients. SARS-CoV-2 antibodies bound to mast cells may be involved in MIS-C and multisystem inflammatory syndrome in adults (MIS-A) following initial COVID-19 infection. SARS-CoV-2 antibodies bound to Fc receptors on macrophages and mast cells may represent two different mechanisms for ADE in patients. These two different ADE risks have possible implications for SARS-CoV-2 B-cell vaccines for subsets of populations based on age, cross-reactive antibodies, variabilities in antibody levels over time, and pregnancy. These models place increased emphasis on the importance of developing safe SARS-CoV-2 T cell vaccines that are not dependent upon antibodies.
Collapse
Affiliation(s)
- Darrell O. Ricke
- Biological and Chemical Technologies, Massachusetts Institute of Technology Lincoln Laboratory, Biotechnology and Human Systems, Lexington, MA, United States
| |
Collapse
|
17
|
Daou A. COVID-19 Vaccination: From Interesting Agent to the Patient. Vaccines (Basel) 2021; 9:120. [PMID: 33546347 PMCID: PMC7913564 DOI: 10.3390/vaccines9020120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/18/2021] [Accepted: 01/21/2021] [Indexed: 12/21/2022] Open
Abstract
The vaccination for the novel Coronavirus (COVID-19) is undergoing its final stages of analysis and testing. It is an impressive feat under the circumstances that we are on the verge of a potential breakthrough vaccination. This will help reduce the stress for millions of people around the globe, helping to restore worldwide normalcy. In this review, the analysis looks into how the new branch of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) came into the forefront of the world like a pandemic. This review will break down the details of what COVID-19 is, the viral family it belongs to and its background of how this family of viruses alters bodily functions by attacking vital human respiratory organs, the circulatory system, the central nervous system and the gastrointestinal tract. This review also looks at the process a new drug analogue undergoes, from (i) being a promising lead compound to (ii) being released into the market, from the drug development and discovery stage right through to FDA approval and aftermarket research. This review also addresses viable reasoning as to why the SARS-CoV-2 vaccine may have taken much less time than normal in order for it to be released for use.
Collapse
Affiliation(s)
- Anis Daou
- Pharmaceutical Sciences Department, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
18
|
Egieyeh S, Egieyeh E, Malan S, Christofells A, Fielding B. Computational drug repurposing strategy predicted peptide-based drugs that can potentially inhibit the interaction of SARS-CoV-2 spike protein with its target (humanACE2). PLoS One 2021; 16:e0245258. [PMID: 33417604 PMCID: PMC7793299 DOI: 10.1371/journal.pone.0245258] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/26/2020] [Indexed: 01/24/2023] Open
Abstract
Drug repurposing for COVID-19 has several potential benefits including shorter development time, reduced costs and regulatory support for faster time to market for treatment that can alleviate the current pandemic. The current study used molecular docking, molecular dynamics and protein-protein interaction simulations to predict drugs from the Drug Bank that can bind to the SARS-CoV-2 spike protein interacting surface on the human angiotensin-converting enzyme 2 (hACE2) receptor. The study predicted a number of peptide-based drugs, including Sar9 Met (O2)11-Substance P and BV2, that might bind sufficiently to the hACE2 receptor to modulate the protein-protein interaction required for infection by the SARS-CoV-2 virus. Such drugs could be validated in vitro or in vivo as potential inhibitors of the interaction of SARS-CoV-2 spike protein with the human angiotensin-converting enzyme 2 (hACE2) in the airway. Exploration of the proposed and current pharmacological indications of the peptide drugs predicted as potential inhibitors of the interaction between the spike protein and hACE2 receptor revealed that some of the predicted peptide drugs have been investigated for the treatment of acute respiratory distress syndrome (ARDS), viral infection, inflammation and angioedema, and to stimulate the immune system, and potentiate antiviral agents against influenza virus. Furthermore, these predicted drug hits may be used as a basis to design new peptide or peptidomimetic drugs with better affinity and specificity for the hACE2 receptor that may prevent interaction between SARS-CoV-2 spike protein and hACE2 that is prerequisite to the infection by the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Samuel Egieyeh
- Computational Pharmacology and Cheminformatics Research Group, Pharmacology and Clinical Pharmacy Unit, School of Pharmacy, University of the Western Cape, Cape Town, South Africa
- * E-mail:
| | - Elizabeth Egieyeh
- Pharmacology and Clinical Pharmacy Unit, School of Pharmacy, University of the Western Cape, Cape Town, South Africa
| | - Sarel Malan
- Pharmaceutical Chemistry Unit, School of Pharmacy, University of the Western Cape, Cape Town, South Africa
| | - Alan Christofells
- South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Cape Town, South Africa
| | - Burtram Fielding
- Molecular Biology and Virology Research Laboratory, Department of Medical Biosciences, University of the Western Cape, Cape Town, South Africa
| |
Collapse
|
19
|
Soleimanpour S, Yaghoubi A. COVID-19 vaccine: where are we now and where should we go? Expert Rev Vaccines 2021; 20:23-44. [PMID: 33435774 PMCID: PMC7898300 DOI: 10.1080/14760584.2021.1875824] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 01/11/2021] [Indexed: 01/12/2023]
Abstract
INTRODUCTION The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has currently caused the pandemic with a high progressive speed and has been considered as the global public health crisis in 2020. This new member of the coronavirus family has created a potentially fatal disease, called coronavirus disease-2019 (COVID-19). Despite the continuous efforts of researchers to find effective vaccines and drugs for COVID-19, there is still no success in this matter. AREAS COVERED Here, the literature regarding the COVID-19 vaccine candidates currently in the clinical trials, as well as main candidates in pre-clinical stages for development and research, were reviewed. These candidates have been developed under five different major platforms, including live-attenuated vaccine, mRNA-based vaccine, DNA vaccines, inactivated virus, and viral-vector-based vaccine. EXPERT OPINION There are several limitations in the field of the rapid vaccine development against SARS-CoV-2, and other members of the coronavirus family such as SARS-CoV and MERS-CoV. The key challenges of designing an effective vaccine within a short time include finding the virulence ability of an emerging virus and potential antigen, choosing suitable experimental models and efficient route of administration, the immune-response study, designing the clinical trials, and determining the safety, as well as efficacy.
Collapse
Affiliation(s)
- Saman Soleimanpour
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atieh Yaghoubi
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Ladner JT, Henson SN, Boyle AS, Engelbrektson AL, Fink ZW, Rahee F, D'ambrozio J, Schaecher KE, Stone M, Dong W, Dadwal S, Yu J, Caligiuri MA, Cieplak P, Bjørås M, Fenstad MH, Nordbø SA, Kainov DE, Muranaka N, Chee MS, Shiryaev SA, Altin JA. Epitope-resolved profiling of the SARS-CoV-2 antibody response identifies cross-reactivity with endemic human coronaviruses. CELL REPORTS MEDICINE 2021; 2:100189. [PMID: 33495758 PMCID: PMC7816965 DOI: 10.1016/j.xcrm.2020.100189] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/11/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022]
Abstract
The SARS-CoV-2 proteome shares regions of conservation with endemic human coronaviruses (CoVs), but it remains unknown to what extent these may be cross-recognized by the antibody response. Here, we study cross-reactivity using a highly multiplexed peptide assay (PepSeq) to generate an epitope-resolved view of IgG reactivity across all human CoVs in both COVID-19 convalescent and negative donors. PepSeq resolves epitopes across the SARS-CoV-2 Spike and Nucleocapsid proteins that are commonly targeted in convalescent donors, including several sites also recognized in some uninfected controls. By comparing patterns of homologous reactivity between CoVs and using targeted antibody-depletion experiments, we demonstrate that SARS-CoV-2 elicits antibodies that cross-recognize pandemic and endemic CoV antigens at two Spike S2 subunit epitopes. We further show that these cross-reactive antibodies preferentially bind endemic homologs. Our findings highlight sites at which the SARS-CoV-2 response appears to be shaped by previous CoV exposures and which have the potential to raise broadly neutralizing responses. PepSeq enables fully in vitro, highly multiplexed peptide-based antibody assays Epitope mapping shows preexisting antibody reactivity to SARS-CoV-2 antigens Antibodies cross-recognize endemic and pandemic antigens in the Spike S2 subunit Cross-reactive antibodies raised by SARS-CoV-2 preferentially bind endemic homologs
Collapse
Affiliation(s)
- Jason T Ladner
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Sierra N Henson
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | - Annalee S Boyle
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | - Anna L Engelbrektson
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | - Zane W Fink
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Fatima Rahee
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | | | | | - Mars Stone
- Vitalant Research Institute and Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Wenjuan Dong
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Sanjeet Dadwal
- Division of Infectious Diseases, City of Hope National Medical Center, Duarte, CA, USA
| | - Jianhua Yu
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Michael A Caligiuri
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Piotr Cieplak
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Magnar Bjørås
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mona H Fenstad
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Immunology and Transfusion Medicine, St. Olavs Hospital, Trondheim, Norway
| | - Svein A Nordbø
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Medical Microbiology, St. Olavs Hospital, Trondheim, Norway
| | - Denis E Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | - Sergey A Shiryaev
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - John A Altin
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| |
Collapse
|
21
|
Zhang XY, Guo J, Wan X, Zhou JG, Jin WP, Lu J, Wang WH, Yang AN, Liu DX, Shi ZL, Yuan ZM, Li XG, Meng SL, Duan K, Wang ZJ, Yang XM, Shen S. Biochemical and antigenic characterization of the structural proteins and their post-translational modifications in purified SARS-CoV-2 virions of an inactivated vaccine candidate. Emerg Microbes Infect 2020; 9:2653-2662. [PMID: 33232205 PMCID: PMC7738289 DOI: 10.1080/22221751.2020.1855945] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the face of COVID-19 pandemic caused by the newly emerged SARS-CoV-2, an inactivated, Vero cell-based, whole virion vaccine candidate has been developed and entered into phase III clinical trials within six months. Biochemical and immunogenic characterization of structural proteins and their post-translational modifications in virions, the end-products of the vaccine candidate, would be essential for the quality control and process development of vaccine products and for studying the immunogenicity and pathogenesis of SARS-CoV-2. By using a panel of rabbit antisera against virions and five structural proteins together with a convalescent serum, the spike (S) glycoprotein was shown to be N-linked glycosylated, PNGase F-sensitive, endoglycosidase H-resistant and cleaved by Furin-like proteases into S1 and S2 subunits. The full-length S and S1/S2 subunits could form homodimers/trimers. The membrane (M) protein was partially N-linked glycosylated; the accessory protein 3a existed in three different forms, indicative of cleavage and dimerization. Furthermore, analysis of the antigenicity of these proteins and their post-translationally modified forms demonstrated that S protein induced the strongest antibody response in both convalescent and immunized animal sera. Interestingly, immunization with the inactivated vaccine did not elicit antibody response against the S2 subunit, whereas strong antibody response against both S1 and S2 subunits was detected in the convalescent serum. Moreover, vaccination stimulated stronger antibody response against S multimers than did the natural infection. This study revealed that the native S glycoprotein stimulated neutralizing antibodies, while bacterially-expressed S fragments did not. The study on S modifications would facilitate design of S-based anti-SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Xiao-Yu Zhang
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Jing Guo
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Xin Wan
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Jin-Ge Zhou
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Wei-Ping Jin
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Jia Lu
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Wen-Hui Wang
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - An-Na Yang
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Ding Xiang Liu
- South China Agricultural University, Guangdong Province Key Laboratory Microbial Signals & Disease Co, & Integrative Microbiology Research Center, Guangzhou, People's Republic of China
| | - Zheng-Li Shi
- Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Zhi-Ming Yuan
- Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Xin-Guo Li
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Sheng-Li Meng
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Kai Duan
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Ze-Jun Wang
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Xiao-Ming Yang
- China National Biotech Group Company Ltd, Beijing, People's Republic of China
| | - Shuo Shen
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| |
Collapse
|
22
|
Cimolai N. Applying Immune Instincts and Maternal Intelligence from Comparative Microbiology to COVID-19. ACTA ACUST UNITED AC 2020; 2:2670-2683. [PMID: 33195997 PMCID: PMC7652409 DOI: 10.1007/s42399-020-00634-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2020] [Indexed: 01/02/2023]
Abstract
New data specific to COVID-19 are emerging quickly on key issues of immunity and prevention, but past research in coronavirology and for other human pathogens (e.g., Mycoplasma pneumoniae) has been available and of great relevance. Considerable study of endemic human coronaviruses has shown that neutralizing antibody correlates with protection, but effective clinical protection is variable for subsequent virus exposure. Animal coronavirus research has emphasized the importance of local mucosal protection (especially IgA) and systemic responses. Animal model and human post-infection studies for SARS-CoV and MERS-CoV are largely corroborative. Whether for passive therapeutic strategies or vaccination, these findings provide a template for COVID-19. Many approaches to vaccination have emerged, and there may be more than one vaccine that will be applied, but individualized obstacles and concerns for administration, efficacy, and safety are inevitable. Regardless of safeguards or promises that may be understood from laboratory or vertebrate experiments, observations from large-scale human trials will ultimately prove to shape the medical future. Focus on common mucosal immunity can be underrated, and equally or more, focus on lactogenic immunity may be underestimated. In understanding both passive immunity and protection, the body is already primed to educate us with decisions of what constitutes protection and harm. This review provides key insights that drive hypotheses into how the instinct of immunity and the intelligence of the maternal component of the common mucosal immune system has already guided us and may continue to do so effectively into a bright and safe future.
Collapse
Affiliation(s)
- Nevio Cimolai
- Faculty of Medicine, The University of British Columbia, Vancouver, BC Canada
- Children’s and Women’s Health Centre of British Columbia, 4480 Oak Street, Vancouver, BC V6H3V4 Canada
| |
Collapse
|
23
|
Tregoning JS, Brown ES, Cheeseman HM, Flight KE, Higham SL, Lemm N, Pierce BF, Stirling DC, Wang Z, Pollock KM. Vaccines for COVID-19. Clin Exp Immunol 2020; 202:162-192. [PMID: 32935331 PMCID: PMC7597597 DOI: 10.1111/cei.13517] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
Since the emergence of COVID-19, caused by the SARS-CoV-2 virus at the end of 2019, there has been an explosion of vaccine development. By 24 September 2020, a staggering number of vaccines (more than 200) had started preclinical development, of which 43 had entered clinical trials, including some approaches that have not previously been licensed for human vaccines. Vaccines have been widely considered as part of the exit strategy to enable the return to previous patterns of working, schooling and socializing. Importantly, to effectively control the COVID-19 pandemic, production needs to be scaled-up from a small number of preclinical doses to enough filled vials to immunize the world's population, which requires close engagement with manufacturers and regulators. It will require a global effort to control the virus, necessitating equitable access for all countries to effective vaccines. This review explores the immune responses required to protect against SARS-CoV-2 and the potential for vaccine-induced immunopathology. We describe the profile of the different platforms and the advantages and disadvantages of each approach. The review also addresses the critical steps between promising preclinical leads and manufacturing at scale. The issues faced during this pandemic and the platforms being developed to address it will be invaluable for future outbreak control. Nine months after the outbreak began we are at a point where preclinical and early clinical data are being generated for the vaccines; an overview of this important area will help our understanding of the next phases.
Collapse
Affiliation(s)
- J. S. Tregoning
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - E. S. Brown
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - H. M. Cheeseman
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - K. E. Flight
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - S. L. Higham
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - N.‐M. Lemm
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - B. F. Pierce
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - D. C. Stirling
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - Z. Wang
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| | - K. M. Pollock
- Department of Infectious DiseaseSt Mary’s CampusImperial College LondonLondonUK
| |
Collapse
|
24
|
Structural Basis of SARS-CoV-2 and SARS-CoV Antibody Interactions. Trends Immunol 2020; 41:1006-1022. [PMID: 33041212 PMCID: PMC7498231 DOI: 10.1016/j.it.2020.09.004] [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: 06/05/2020] [Revised: 09/12/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022]
Abstract
The 2019 coronavirus pandemic remains a major public health concern. Neutralizing antibodies (nAbs) represent a cutting-edge antiviral strategy. We focus here on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and SARS-CoV, and discuss current progress in antibody research against rampant SARS-CoV-2 infections. We provide a perspective on the mechanisms of SARS-CoV-2-derived nAbs, comparing these with existing SARS-CoV-derived antibodies. We offer insight into how these antibodies cross-react and cross-neutralize by analyzing available structures of spike (S) glycoprotein-antibody complexes. We also propose ways of adopting antibody-based strategies - such as cocktail antibody therapeutics against SARS-CoV-2 - to overcome the possible resistance of currently identified mutants and mitigate possible antibody-dependent enhancement (ADE) pathologies. This review provides a platform for the progression of antibody and vaccine design against SARS-CoV-2, and possibly against future coronavirus pandemics.
Collapse
|
25
|
Meyer B, Torriani G, Yerly S, Mazza L, Calame A, Arm-Vernez I, Zimmer G, Agoritsas T, Stirnemann J, Spechbach H, Guessous I, Stringhini S, Pugin J, Roux-Lombard P, Fontao L, Siegrist CA, Eckerle I, Vuilleumier N, Kaiser L. Validation of a commercially available SARS-CoV-2 serological immunoassay. Clin Microbiol Infect 2020; 26:1386-1394. [PMID: 32603801 PMCID: PMC7320699 DOI: 10.1016/j.cmi.2020.06.024] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 01/20/2023]
Abstract
OBJECTIVES To validate the diagnostic accuracy of a Euroimmun SARS-CoV-2 IgG and IgA immunoassay for COVID-19. METHODS In this unmatched (1:2) case-control validation study, we used sera of 181 laboratory-confirmed SARS-CoV-2 cases and 326 controls collected before SARS-CoV-2 emergence. Diagnostic accuracy of the immunoassay was assessed against a whole spike protein-based recombinant immunofluorescence assay (rIFA) by receiver operating characteristic (ROC) analyses. Discrepant cases between ELISA and rIFA were further tested by pseudo-neutralization assay. RESULTS COVID-19 patients were more likely to be male and older than controls, and 50.3% were hospitalized. ROC curve analyses indicated that IgG and IgA had high diagnostic accuracies with AUCs of 0.990 (95% Confidence Interval [95%CI]: 0.983-0.996) and 0.978 (95%CI: 0.967-0.989), respectively. IgG assays outperformed IgA assays (p=0.01). Taking an assessed 15% inter-assay imprecision into account, an optimized IgG ratio cut-off > 2.5 displayed a 100% specificity (95%CI: 99-100) and a 100% positive predictive value (95%CI: 96-100). A 0.8 cut-off displayed a 94% sensitivity (95%CI: 88-97) and a 97% negative predictive value (95%CI: 95-99). Substituting the upper threshold for the manufacturer's, improved assay performance, leaving 8.9% of IgG ratios indeterminate between 0.8-2.5. CONCLUSIONS The Euroimmun assay displays a nearly optimal diagnostic accuracy using IgG against SARS-CoV-2 in patient samples, with no obvious gains from IgA serology. The optimized cut-offs are fit for rule-in and rule-out purposes, allowing determination of whether individuals in our study population have been exposed to SARS-CoV-2 or not. IgG serology should however not be considered as a surrogate of protection at this stage.
Collapse
Affiliation(s)
- B Meyer
- Centre for Vaccinology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - G Torriani
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - S Yerly
- Laboratory of Virology, Geneva University Hospitals, Geneva, Switzerland
| | - L Mazza
- Laboratory of Virology, Geneva University Hospitals, Geneva, Switzerland
| | - A Calame
- Division of Infectious Disease, Geneva University Hospitals, Geneva, Switzerland
| | - I Arm-Vernez
- Laboratory of Virology, Geneva University Hospitals, Geneva, Switzerland
| | - G Zimmer
- Institute of Virology and Immunology (IVI), Mittelhäusern, Switzerland; Department of Infectious Diseases and Pathobiology (DIP), Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - T Agoritsas
- Division of General Internal Medicine, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland; Department of Health Research Methods, Evidence, and Impact, Hamilton, Ontario, Canada
| | - J Stirnemann
- Division of General Internal Medicine, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - H Spechbach
- Division and Department of Primary Care Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - I Guessous
- Division and Department of Primary Care Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - S Stringhini
- Division and Department of Primary Care Medicine, Geneva University Hospitals, Geneva, Switzerland; Unit of Population Epidemiology, Division of Primary Care, Geneva University Hospitals, Geneva, Switzerland
| | - J Pugin
- Division of Intensive Care, Geneva University Hospitals, Geneva, Switzerland
| | - P Roux-Lombard
- Division of Laboratory Medicine, Department of Diagnostics, Geneva University Hospitals and Geneva University, Geneva, Switzerland
| | - L Fontao
- Division of Dermatology and of Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - C-A Siegrist
- Centre for Vaccinology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - I Eckerle
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland; Division of Infectious Disease, Geneva University Hospitals, Geneva, Switzerland; Geneva Centre for Emerging Viral Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - N Vuilleumier
- Division of Laboratory Medicine, Department of Diagnostics, Geneva University Hospitals and Geneva University, Geneva, Switzerland; Division of Laboratory Medicine, Department of Medicine, Faculty of Medicine, Geneva, Switzerland
| | - L Kaiser
- Laboratory of Virology, Geneva University Hospitals, Geneva, Switzerland; Division of Infectious Disease, Geneva University Hospitals, Geneva, Switzerland; Geneva Centre for Emerging Viral Diseases, Geneva University Hospitals, Geneva, Switzerland.
| |
Collapse
|
26
|
Tong PBV, Lin LY, Tran TH. Coronaviruses pandemics: Can neutralizing antibodies help? Life Sci 2020; 255:117836. [PMID: 32450171 PMCID: PMC7243778 DOI: 10.1016/j.lfs.2020.117836] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
Abstract
For the first time in Homo sapiens history, possibly, most of human activities is stopped by coronavirus disease 2019 (COVID-19). Nearly eight billion people of this world are facing a great challenge, maybe not "to be or not to be" yet, but unpredictable. What happens to other major pandemics in the past, and how human beings went through these hurdles? The human body is equipped with the immune system that can recognize, respond and fight against pathogens such as viruses. Following the innate response, immune system processes the adaptive response by which each pathogen is encoded and recorded in memory system. The humoral reaction containing cytokines and antibodies is expected to activate when the pathogens come back. Exploiting this nature of body protection, neutralizing antibodies have been investigated. Learning from past, in parallel to SARS-CoV-2, other coronaviruses SARS-CoV and MERS-CoV who caused previous pandemics, are recalled in this review. We here propose insights of origin and characteristics and perspective for the future of antibodies development.
Collapse
Affiliation(s)
- Phuoc-Bao-Viet Tong
- INSERM U1109, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Li-Yun Lin
- INSERM U1109, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi 12116, Viet Nam; PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, No.167 Hoang Ngan, Trung Hoa, Cau Giay, Hanoi 11313, Viet Nam.
| |
Collapse
|
27
|
Zheng Z, Monteil VM, Maurer-Stroh S, Yew CW, Leong C, Mohd-Ismail NK, Cheyyatraivendran Arularasu S, Chow VTK, Lin RTP, Mirazimi A, Hong W, Tan YJ. Monoclonal antibodies for the S2 subunit of spike of SARS-CoV-1 cross-react with the newly-emerged SARS-CoV-2. ACTA ACUST UNITED AC 2020; 25. [PMID: 32700671 PMCID: PMC7376845 DOI: 10.2807/1560-7917.es.2020.25.28.2000291] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background A novel coronavirus, SARS-CoV-2, which emerged at the end of 2019 and causes COVID-19, has resulted in worldwide human infections. While genetically distinct, SARS-CoV-1, the aetiological agent responsible for an outbreak of severe acute respiratory syndrome (SARS) in 2002–2003, utilises the same host cell receptor as SARS-CoV-2 for entry: angiotensin-converting enzyme 2 (ACE2). Parts of the SARS-CoV-1 spike glycoprotein (S protein), which interacts with ACE2, appear conserved in SARS-CoV-2. Aim The cross-reactivity with SARS-CoV-2 of monoclonal antibodies (mAbs) previously generated against the S protein of SARS-CoV-1 was assessed. Methods The SARS-CoV-2 S protein sequence was aligned to those of SARS-CoV-1, Middle East respiratory syndrome (MERS) and common-cold coronaviruses. Abilities of mAbs generated against SARS-CoV-1 S protein to bind SARS-CoV-2 or its S protein were tested with SARS-CoV-2 infected cells as well as cells expressing either the full length protein or a fragment of its S2 subunit. Quantitative ELISA was also performed to compare binding of mAbs to recombinant S protein. Results An immunogenic domain in the S2 subunit of SARS-CoV-1 S protein is highly conserved in SARS-CoV-2 but not in MERS and human common-cold coronaviruses. Four murine mAbs raised against this immunogenic fragment could recognise SARS-CoV-2 S protein expressed in mammalian cell lines. In particular, mAb 1A9 was demonstrated to detect S protein in SARS-CoV-2-infected cells and is suitable for use in a sandwich ELISA format. Conclusion The cross-reactive mAbs may serve as useful tools for SARS-CoV-2 research and for the development of diagnostic assays for COVID-19.
Collapse
Affiliation(s)
- Zhiqiang Zheng
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore.,Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Vanessa Marthe Monteil
- Public Health Agency of Sweden, Stockholm, Sweden.,Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
| | - Sebastian Maurer-Stroh
- National Public Health Laboratory (NPHL), National Centre for Infectious Diseases (NCID), Singapore.,Department of Biological Sciences (DBS), National University of Singapore, Singapore.,Bioinformatics Institute (BII), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Chow Wenn Yew
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Carol Leong
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Nur Khairiah Mohd-Ismail
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore.,Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Suganya Cheyyatraivendran Arularasu
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore.,Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Vincent Tak Kwong Chow
- Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Raymond Tzer Pin Lin
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore.,National Public Health Laboratory (NPHL), National Centre for Infectious Diseases (NCID), Singapore
| | - Ali Mirazimi
- National Veterinary Institute, Uppsala, Sweden.,Public Health Agency of Sweden, Stockholm, Sweden.,Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
| | - Wanjin Hong
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Yee-Joo Tan
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore.,Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore.,Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| |
Collapse
|
28
|
Ladner JT, Henson SN, Boyle AS, Engelbrektson AL, Fink ZW, Rahee F, D’ambrozio J, Schaecher KE, Stone M, Dong W, Dadwal S, Yu J, Caligiuri MA, Cieplak P, Bjørås M, Fenstad MH, Nordbø SA, Kainov DE, Muranaka N, Chee MS, Shiryaev SA, Altin JA. Epitope-resolved profiling of the SARS-CoV-2 antibody response identifies cross-reactivity with an endemic human CoV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.27.222943. [PMID: 32743570 PMCID: PMC7386487 DOI: 10.1101/2020.07.27.222943] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A high-resolution understanding of the antibody response to SARS-CoV-2 is important for the design of effective diagnostics, vaccines and therapeutics. However, SARS-CoV-2 antibody epitopes remain largely uncharacterized, and it is unknown whether and how the response may cross-react with related viruses. Here, we use a multiplexed peptide assay ('PepSeq') to generate an epitope-resolved view of reactivity across all human coronaviruses. PepSeq accurately detects SARS-CoV-2 exposure and resolves epitopes across the Spike and Nucleocapsid proteins. Two of these represent recurrent reactivities to conserved, functionally-important sites in the Spike S2 subunit, regions that we show are also targeted for the endemic coronaviruses in pre-pandemic controls. At one of these sites, we demonstrate that the SARS-CoV-2 response strongly and recurrently cross-reacts with the endemic virus hCoV-OC43. Our analyses reveal new diagnostic and therapeutic targets, including a site at which SARS-CoV-2 may recruit common pre-existing antibodies and with the potential for broadly-neutralizing responses.
Collapse
Affiliation(s)
- Jason T Ladner
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Sierra N Henson
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | - Annalee S Boyle
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | - Anna L Engelbrektson
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | - Zane W Fink
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Fatima Rahee
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| | | | | | - Mars Stone
- Vitalant Research Institute, San Francisco, CA, USA
| | - Wenjuan Dong
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Sanjeet Dadwal
- Division of Infectious Diseases, City of Hope National Medical Center, Duarte, CA, USA
| | - Jianhua Yu
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Michael A Caligiuri
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Piotr Cieplak
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Magnar Bjørås
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mona H Fenstad
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Immunology and Transfusion Medicine, St. Olavs Hospital, Trondheim, Norway
| | - Svein A Nordbø
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Medical Microbiology, St. Olavs Hospital, Trondheim, Norway
| | - Denis E Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | - Sergey A Shiryaev
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - John A Altin
- The Translational Genomics Research Institute (TGen), Phoenix and Flagstaff, AZ, USA
| |
Collapse
|
29
|
Protective Immunity against SARS Subunit Vaccine Candidates Based on Spike Protein: Lessons for Coronavirus Vaccine Development. J Immunol Res 2020; 2020:7201752. [PMID: 32695833 PMCID: PMC7368938 DOI: 10.1155/2020/7201752] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 06/09/2020] [Indexed: 12/21/2022] Open
Abstract
The recent outbreak of the novel coronavirus disease, COVID-19, has highlighted the threat that highly pathogenic coronaviruses have on global health security and the imminent need to design an effective vaccine for prevention purposes. Although several attempts have been made to develop vaccines against human coronavirus infections since the emergence of Severe Acute Respiratory Syndrome coronavirus (SARS-CoV) in 2003, there is no available licensed vaccine yet. A better understanding of previous coronavirus vaccine studies may help to design a vaccine for the newly emerged virus, SARS-CoV-2, that may also cover other pathogenic coronaviruses as a potentially universal vaccine. In general, coronavirus spike protein is the major antigen for the vaccine design as it can induce neutralizing antibodies and protective immunity. By considering the high genetic similarity between SARS-CoV and SARS-CoV-2, here, protective immunity against SARS-CoV spike subunit vaccine candidates in animal models has been reviewed to gain advances that can facilitate coronavirus vaccine development in the near future.
Collapse
|
30
|
Zeidler A, Karpinski TM. SARS-CoV, MERS-CoV, SARS-CoV-2 Comparison of Three Emerging Coronaviruses. Jundishapur J Microbiol 2020; 13. [DOI: 10.5812/jjm.103744] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 08/29/2023] Open
Abstract
: In December 2019, in Wuhan, China began the outbreak of the new severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) epidemic. As a result of rapid spread, it turned into a pandemic announced by WHO on March 11, 2020. SARS-CoV-2 is an etiological factor of a new disease called COVID-19. The virus is transmitted mainly through the droplet route. In most cases, it causes mild symptoms such as fever, dry cough, weakness, and muscle pain; less common symptoms include sore throat, runny nose, diarrhea, and chills. However, among people with impaired immunity and comorbidities, as well as among older people, it leads to life-threatening complications in the form of acute respiratory distress syndrome (ARDS), sepsis, and septic shock. Moreover, SARS-CoV-2 is the third highly pathogenic in humans and easily spreading coronavirus after the virus of a severe acute respiratory syndrome (SARS) in 2002 - 2003 and virus of the Middle East respiratory syndrome (MERS) in 2012. This review summarizes current information on the emergence, origin, diversity, and common characteristics, as well as the epidemiology of the above three highly contagious coronaviruses.
Collapse
|
31
|
Zheng Z, Monteil VM, Maurer-Stroh S, Yew CW, Leong C, Mohd-Ismail NK, Cheyyatraivendran Arularasu S, Chow VTK, Lin RTP, Mirazimi A, Hong W, Tan YJ. Monoclonal antibodies for the S2 subunit of spike of SARS-CoV-1 cross-react with the newly-emerged SARS-CoV-2. Euro Surveill 2020. [PMID: 32700671 DOI: 10.1101/2020.03.06.980037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
BackgroundA novel coronavirus, SARS-CoV-2, which emerged at the end of 2019 and causes COVID-19, has resulted in worldwide human infections. While genetically distinct, SARS-CoV-1, the aetiological agent responsible for an outbreak of severe acute respiratory syndrome (SARS) in 2002-2003, utilises the same host cell receptor as SARS-CoV-2 for entry: angiotensin-converting enzyme 2 (ACE2). Parts of the SARS-CoV-1 spike glycoprotein (S protein), which interacts with ACE2, appear conserved in SARS-CoV-2.AimThe cross-reactivity with SARS-CoV-2 of monoclonal antibodies (mAbs) previously generated against the S protein of SARS-CoV-1 was assessed.MethodsThe SARS-CoV-2 S protein sequence was aligned to those of SARS-CoV-1, Middle East respiratory syndrome (MERS) and common-cold coronaviruses. Abilities of mAbs generated against SARS-CoV-1 S protein to bind SARS-CoV-2 or its S protein were tested with SARS-CoV-2 infected cells as well as cells expressing either the full length protein or a fragment of its S2 subunit. Quantitative ELISA was also performed to compare binding of mAbs to recombinant S protein.ResultsAn immunogenic domain in the S2 subunit of SARS-CoV-1 S protein is highly conserved in SARS-CoV-2 but not in MERS and human common-cold coronaviruses. Four murine mAbs raised against this immunogenic fragment could recognise SARS-CoV-2 S protein expressed in mammalian cell lines. In particular, mAb 1A9 was demonstrated to detect S protein in SARS-CoV-2-infected cells and is suitable for use in a sandwich ELISA format.ConclusionThe cross-reactive mAbs may serve as useful tools for SARS-CoV-2 research and for the development of diagnostic assays for COVID-19.
Collapse
Affiliation(s)
- Zhiqiang Zheng
- Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Vanessa Marthe Monteil
- Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
- Public Health Agency of Sweden, Stockholm, Sweden
| | - Sebastian Maurer-Stroh
- Bioinformatics Institute (BII), A*STAR (Agency for Science, Technology and Research), Singapore
- Department of Biological Sciences (DBS), National University of Singapore, Singapore
- National Public Health Laboratory (NPHL), National Centre for Infectious Diseases (NCID), Singapore
| | - Chow Wenn Yew
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Carol Leong
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Nur Khairiah Mohd-Ismail
- Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Suganya Cheyyatraivendran Arularasu
- Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Vincent Tak Kwong Chow
- Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Raymond Tzer Pin Lin
- National Public Health Laboratory (NPHL), National Centre for Infectious Diseases (NCID), Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Ali Mirazimi
- National Veterinary Institute, Uppsala, Sweden
- Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
- Public Health Agency of Sweden, Stockholm, Sweden
| | - Wanjin Hong
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Yee-Joo Tan
- Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| |
Collapse
|
32
|
He C, Qin M, Sun X. Highly pathogenic coronaviruses: thrusting vaccine development in the spotlight. Acta Pharm Sin B 2020; 10:1175-1191. [PMID: 32834948 PMCID: PMC7260574 DOI: 10.1016/j.apsb.2020.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 01/03/2023] Open
Abstract
Coronaviruses (CoVs) are a large family of viruses that cause illness ranging from the common cold to more severe diseases such as Severe Acute Respiratory Syndrome (SARS) and Middle East Respiratory Syndrome (MERS). Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) disease (COVID-19) has caused major public health crises. There have been more than 4,400,000 reported cases of COVID-2019 and more than 300,000 reported deaths to date (16/05/2020). SARS-CoV, MERS-CoV and SARS-CoV-2 have attracted widespread global attention due to their high infectivity and pathogenicity. To date, there is no specific treatment proven effective against these viral infectious diseases. Vaccination is considered one of the most effective strategies to prevent viral infections. Therefore, the development of effective vaccines against highly pathogenic coronaviruses is essential. In this review, we will briefly describe coronavirus vaccine design targets, summarize recent advances in the development of coronavirus vaccines, and highlight current adjuvants for improving the efficacy of coronavirus vaccines.
Collapse
|
33
|
SARS-CoV-2: An Update on Potential Antivirals in Light of SARS-CoV Antiviral Drug Discoveries. Vaccines (Basel) 2020; 8:vaccines8020335. [PMID: 32585913 PMCID: PMC7350231 DOI: 10.3390/vaccines8020335] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/04/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Coronaviruses (CoVs) are a group of RNA viruses that are associated with different diseases in animals, birds, and humans. Human CoVs (HCoVs) have long been known to be the causative agents of mild respiratory illnesses. However, two HCoVs associated with severe respiratory diseases are Severe Acute Respiratory Syndrome-CoV (SARS-CoV) and Middle East Respiratory Syndrome-CoV (MERS-CoV). Both viruses resulted in hundreds of deaths after spreading to several countries. Most recently, SARS-CoV-2 has emerged as the third HCoV causing severe respiratory distress syndrome and viral pneumonia (known as COVID-19) in patients from Wuhan, China, in December 2019. Soon after its discovery, SARS-CoV-2 spread to all countries, resulting in millions of cases and thousands of deaths. Since the emergence of SARS-CoV, many research groups have dedicated their resources to discovering effective antivirals that can treat such life-threatening infections. The rapid spread and high fatality rate of SARS-CoV-2 necessitate the quick discovery of effective antivirals to control this outbreak. Since SARS-CoV-2 shares 79% sequence identity with SARS-CoV, several anti-SARS-CoV drugs have shown promise in limiting SARS-CoV-2 replication in vitro and in vivo. In this review, we discuss antivirals described for SARS-CoV and provide an update on therapeutic strategies and antivirals against SARS-CoV-2. The control of the current outbreak will strongly depend on the discovery of effective and safe anti-SARS-CoV-2 drugs.
Collapse
|
34
|
From SARS to MERS, Thrusting Coronaviruses into the Spotlight. Viruses 2019; 11:v11010059. [PMID: 30646565 PMCID: PMC6357155 DOI: 10.3390/v11010059] [Citation(s) in RCA: 704] [Impact Index Per Article: 117.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 01/03/2019] [Accepted: 01/09/2019] [Indexed: 11/30/2022] Open
Abstract
Coronaviruses (CoVs) have formerly been regarded as relatively harmless respiratory pathogens to humans. However, two outbreaks of severe respiratory tract infection, caused by the severe acute respiratory syndrome coronavirus (SARS-CoV) and the Middle East respiratory syndrome coronavirus (MERS-CoV), as a result of zoonotic CoVs crossing the species barrier, caused high pathogenicity and mortality rates in human populations. This brought CoVs global attention and highlighted the importance of controlling infectious pathogens at international borders. In this review, we focus on our current understanding of the epidemiology, pathogenesis, prevention, and treatment of SARS-CoV and MERS-CoV, as well as provides details on the pivotal structure and function of the spike proteins (S proteins) on the surface of each of these viruses. For building up more suitable animal models, we compare the current animal models recapitulating pathogenesis and summarize the potential role of host receptors contributing to diverse host affinity in various species. We outline the research still needed to fully elucidate the pathogenic mechanism of these viruses, to construct reproducible animal models, and ultimately develop countermeasures to conquer not only SARS-CoV and MERS-CoV, but also these emerging coronaviral diseases.
Collapse
|
35
|
Ng OW, Keng CT, Leung CSW, Peiris JSM, Poon LLM, Tan YJ. Substitution at aspartic acid 1128 in the SARS coronavirus spike glycoprotein mediates escape from a S2 domain-targeting neutralizing monoclonal antibody. PLoS One 2014; 9:e102415. [PMID: 25019613 PMCID: PMC4097068 DOI: 10.1371/journal.pone.0102415] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 06/19/2014] [Indexed: 12/28/2022] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) is the etiological agent for the infectious disease, SARS, which first emerged 10 years ago. SARS-CoV is a zoonotic virus that has crossed the species barriers to infect humans. Bats, which harbour a diverse pool of SARS-like CoVs (SL-CoVs), are believed to be the natural reservoir. The SARS-CoV surface Spike (S) protein is a major antigenic determinant in eliciting neutralizing antibody production during SARS-CoV infection. In our previous work, we showed that a panel of murine monoclonal antibodies (mAbs) that target the S2 subunit of the S protein are capable of neutralizing SARS-CoV infection in vitro (Lip KM et al, J Virol. 2006 Jan; 80(2): 941-50). In this study, we report our findings on the characterization of one of these mAbs, known as 1A9, which binds to the S protein at a novel epitope within the S2 subunit at amino acids 1111-1130. MAb 1A9 is a broadly neutralizing mAb that prevents viral entry mediated by the S proteins of human and civet SARS-CoVs as well as bat SL-CoVs. By generating mutant SARS-CoV that escapes the neutralization by mAb 1A9, the residue D1128 in S was found to be crucial for its interaction with mAb 1A9. S protein containing the substitution of D1128 with alanine (D1128A) exhibited a significant decrease in binding capability to mAb 1A9 compared to wild-type S protein. By using a pseudotyped viral entry assay, it was shown that the D1128A substitution in the escape virus allows it to overcome the viral entry blockage by mAb 1A9. In addition, the D1128A mutation was found to exert no effects on the S protein cell surface expression and incorporation into virion particles, suggesting that the escape virus retains the same viral entry property as the wild-type virus.
Collapse
Affiliation(s)
- Oi-Wing Ng
- Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Choong-Tat Keng
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Cynthia Sau-Wai Leung
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong, China
| | - J. S. Malik Peiris
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Leo Lit Man Poon
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Yee-Joo Tan
- Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore
| |
Collapse
|
36
|
Developments in the Search for Small-Molecule Inhibitors for Treatment of Severe Acute Respiratory Syndrome Coronavirus. Antiviral Res 2014. [DOI: 10.1128/9781555815493.ch12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
37
|
Li J, Ulitzky L, Silberstein E, Taylor DR, Viscidi R. Immunogenicity and protection efficacy of monomeric and trimeric recombinant SARS coronavirus spike protein subunit vaccine candidates. Viral Immunol 2013; 26:126-32. [PMID: 23573979 DOI: 10.1089/vim.2012.0076] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Severe acute respiratory syndrome (SARS) is a newly emerging infectious disease, and an effective vaccine is not available. In this study, we compared the immunogenicity and protection efficacy of recombinant proteins corresponding to different domains of the SARS-coronavirus spike protein. Trimeric recombinant proteins were created by fusing the foldon domain derived from T4 bacteriophage to the carboxy-termini of individual domains of the spike protein. While the full-length ectodomain (S) of the spike protein, the full-length ectodomain fused to foldon (S-foldon), the S1 domain (S1), S1-foldon, and the S2 domain(S2) antigens all elicited comparable antibody titers as measured by ELISA, S-foldon induced a significantly higher titer of neutralizing antibody and S2 protein did not elicit virus neutralizing antibodies. When tested in a mouse virus replication model, all the mice vaccinated with the S1, S1-foldon, S, or S-foldon were completely protected.
Collapse
Affiliation(s)
- Jie Li
- Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | | | | | | | | |
Collapse
|
38
|
Chan KH, Chan JFW, Tse H, Chen H, Lau CCY, Cai JP, Tsang AKL, Xiao X, To KKW, Lau SKP, Woo PCY, Zheng BJ, Wang M, Yuen KY. Cross-reactive antibodies in convalescent SARS patients' sera against the emerging novel human coronavirus EMC (2012) by both immunofluorescent and neutralizing antibody tests. J Infect 2013; 67:130-40. [PMID: 23583636 PMCID: PMC7112694 DOI: 10.1016/j.jinf.2013.03.015] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 03/27/2013] [Accepted: 03/31/2013] [Indexed: 01/29/2023]
Abstract
Objectives A severe acute respiratory syndrome (SARS)-like disease due to a novel betacoronavirus, human coronavirus EMC (HCoV-EMC), has emerged recently. HCoV-EMC is phylogenetically closely related to Tylonycteris-bat-coronavirus-HKU4 and Pipistrellus-bat-coronavirus-HKU5 in Hong Kong. We conducted a seroprevalence study on archived sera from 94 game-food animal handlers at a wild life market, 28 SARS patients, and 152 healthy blood donors in Southern China to assess the zoonotic potential and evidence for intrusion of HCoV-EMC and related viruses into humans. Methods Anti-HCoV-EMC and anti-SARS-CoV antibodies were detected using screening indirect immunofluorescence (IF) and confirmatory neutralizing antibody tests. Results Two (2.1%) animal handlers had IF antibody titer of ≥1:20 against both HCoV-EMC and SARS-CoV with neutralizing antibody titer of <1:10. No blood donor had antibody against either virus. Surprisingly, 17/28 (60.7%) of SARS patients had significant IF antibody titers with 7/28 (25%) having anti-HCoV-EMC neutralizing antibodies at low titers which significantly correlated with that of HCoV-OC43. Bioinformatics analysis demonstrated a significant B-cell epitope overlapping the heptad repeat-2 region of Spike protein. Virulence of SARS-CoV over other betacoronaviruses may boost cross-reactive neutralizing antibodies against other betacoronaviruses. Conclusions Convalescent SARS sera may contain cross-reactive antibodies against other betacoronaviruses and confound seroprevalence study for HCoV-EMC.
Collapse
Affiliation(s)
- Kwok-Hung Chan
- Department of Microbiology, Queen Mary Hospital, The University of Hong Kong, Hong Kong Special Administrative Region
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Elshabrawy HA, Coughlin MM, Baker SC, Prabhakar BS. Human monoclonal antibodies against highly conserved HR1 and HR2 domains of the SARS-CoV spike protein are more broadly neutralizing. PLoS One 2012. [PMID: 23185609 PMCID: PMC3503966 DOI: 10.1371/journal.pone.0050366] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Immune sera from convalescent patients have been shown to be effective in the treatment of patients infected with Severe Acute Respiratory Syndrome Virus (SARS-CoV) making passive immune therapy with human monoclonal antibodies an attractive treatment strategy for SARS. Previously, using Xenomouse (Amgen British Columbia Inc), we produced a panel of neutralizing Human monoclonal antibodies (HmAbs) that could specifically bind to the ectodomain of the SARS-CoV spike (S) glycoprotein. Some of the HmAbs were S1 domain specific, while some were not. In this study, we describe non-S1 binding neutralizing HmAbs that can specifically bind to the conserved S2 domain of the S protein. However, unlike the S1 specific HmAbs, the S2 specific HmAbs can neutralize pseudotyped viruses expressing different S proteins containing receptor binding domain sequences of various clinical isolates. These data indicate that HmAbs which bind to conserved regions of the S protein are more suitable for conferring protection against a wide range of SARS-CoV variants and have implications for generating therapeutic antibodies or subunit vaccines against other enveloped viruses.
Collapse
Affiliation(s)
- Hatem A. Elshabrawy
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Melissa M. Coughlin
- Centers for Disease Control and Prevention, Measles, Mumps, Rubella and Herpes Virus Laboratory Branch, Atlanta, Georgia, United States of America
| | - Susan C. Baker
- Department of Microbiology and Immunology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, United States of America
| | - Bellur S. Prabhakar
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
40
|
Coughlin MM, Prabhakar BS. Neutralizing human monoclonal antibodies to severe acute respiratory syndrome coronavirus: target, mechanism of action, and therapeutic potential. Rev Med Virol 2011; 22:2-17. [PMID: 21905149 PMCID: PMC3256278 DOI: 10.1002/rmv.706] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 07/06/2011] [Accepted: 07/07/2011] [Indexed: 12/12/2022]
Abstract
The emergence of Severe Acute Respiratory Syndrome Coronavirus (SARS‐CoV) led to a rapid response not only to contain the outbreak but also to identify possible therapeutic interventions, including the generation of human monoclonal antibodies (hmAbs). hmAbs may be used therapeutically without the drawbacks of chimeric or animal Abs. Several different methods have been used to generate SARS‐CoV specific neutralizing hmAbs including the immunization of transgenic mice, cloning of small chain variable regions from naïve and convalescent patients, and the immortalization of convalescent B cells. Irrespective of the techniques used, the majority of hmAbs specifically reacted with the receptor binding domain (RBD) of the spike (S) protein and likely prevented receptor binding. However, several hmAbs that can bind to epitopes either within the RBD, located N terminal of the RBD or in the S2 domain, and neutralize the virus with or without inhibiting receptor binding have been identified. Therapeutic utility of hmAbs has been further elucidated through the identification of potential combinations of hmAbs that could neutralize viral variants including escape mutants selected using hmAbs. These results suggest that a cocktail of hmAbs that can bind to unique epitopes and have different mechanisms of action might be of clinical utility against SARS‐CoV infection, and indicate that a similar approach may be applied to treat other viral infections. Copyright © 2011 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Melissa M Coughlin
- Centers for Disease Control and Prevention, Measles, Mumps, Rubella and Herpes Virus Laboratory Branch, Atlanta, GA, USA.
| | | |
Collapse
|
41
|
Chen YN, Wu CC, Lin TL. Identification and characterization of a neutralizing-epitope-containing spike protein fragment in turkey coronavirus. Arch Virol 2011; 156:1525-35. [PMID: 21594597 PMCID: PMC7086772 DOI: 10.1007/s00705-011-1020-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 05/02/2011] [Indexed: 12/27/2022]
Abstract
Little is known about the neutralizing epitopes in turkey coronavirus (TCoV). The spike (S) protein gene of TCoV was divided into 10 fragments to identify the antigenic region containing neutralizing epitopes. The expression and antigenicity of S fragments was confirmed by immunofluorescence antibody (IFA) assay using an anti-histidine monoclonal antibody or anti-TCoV serum. Polyclonal antibodies raised against expressed S1 (amino acid position 1 to 573 from start codon of S protein), 4F/4R (482-678), 6F/6R (830-1071), or Mod4F/Epi4R (476-520) S fragment recognized native S1 protein and TCoV in the intestines of TCoV-infected turkey embryos. Anti-TCoV serum reacted with recombinant 4F/4R, 6F/6R, and Mod4F/Epi4R in a western blot. The results of a virus neutralization assay indicated that the carboxyl terminal region of the S1 protein (Mod4F/Epi4R) or the combined carboxyl terminal S1 and amino terminal S2 protein (4F/4R) possesses the neutralizing epitopes, while the S2 fragment (6F/6R) contains antigenic epitopes but not neutralizing epitopes.
Collapse
Affiliation(s)
- Yi-Ning Chen
- Department of Comparative Pathobiology, Purdue University, 406 South University Street, West Lafayette, IN 47907, USA
| | | | | |
Collapse
|
42
|
Keng CT, Akerström S, Leung CSW, Poon LLM, Peiris JSM, Mirazimi A, Tan YJ. SARS coronavirus 8b reduces viral replication by down-regulating E via an ubiquitin-independent proteasome pathway. Microbes Infect 2010; 13:179-88. [PMID: 21035562 PMCID: PMC7110893 DOI: 10.1016/j.micinf.2010.10.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 10/19/2010] [Accepted: 10/21/2010] [Indexed: 12/27/2022]
Abstract
The severe acute respiratory syndrome coronavirus (SARS-CoV) 8b protein, which is not expressed by other known coronaviruses, can down-regulate the envelope (E) protein via a proteasome-dependent pathway. Here, we showed that the down-regulation of E is not dependent on the lysine residues on 8b and the reduction of polyubiquitination of E mutants is not correlated with their down-regulation by 8b, suggesting an ubiquitin-independent proteasome pathway is involved. A time-course study revealed that 8b was expressed at late-stages of SARS-CoV infection. By using Vero E6 cells stably expressing green fluorescence protein-tagged 8b, ectopic expression of 8b was shown to significantly reduce the production of progeny virus and down-regulate E expression. Taken together, these results suggest that 8b negatively modulates virus replication by down-regulating E via an ubiquitin-independent proteasome pathway.
Collapse
Affiliation(s)
- Choong-Tat Keng
- Collaborative Anti-Viral Research Group, Institute of Molecular and Cell Biology, Singapore
| | | | | | | | | | | | | |
Collapse
|
43
|
Du L, Zhao G, Chan CCS, Li L, He Y, Zhou Y, Zheng BJ, Jiang S. A 219-mer CHO-expressing receptor-binding domain of SARS-CoV S protein induces potent immune responses and protective immunity. Viral Immunol 2010; 23:211-9. [PMID: 20374001 DOI: 10.1089/vim.2009.0090] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Development of vaccines is essential for the prevention of future recurrences of severe acute respiratory syndrome (SARS), caused by the SARS coronavirus (SARS-CoV). The spike (S) protein, especially receptor-binding domain (RBD) of SARS-CoV, plays important roles in the prevention of SARS infection, and is thus an important component in SARS vaccine development. In this study, we expressed a 219-mer (residues 318-536) RBD protein in Chinese hamster ovary (CHO)-K1 cells (RBD219-CHO), and tested its immune responses and protective immunity in a mouse model. The results showed that this recombinant protein was correctly folded, being able to maintain intact conformation and authentic antigenicity. It could induce strong humoral and cellular immune responses and high titers of neutralizing antibodies in the vaccinated mice. RBD219-CHO protein elicited potent protective immunity that protected all vaccinated mice from SARS-CoV challenge. These results suggest that the recombinant RBD219-CHO protein has great potential for the development of an effective and safe SARS subunit vaccine.
Collapse
Affiliation(s)
- Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York 10065, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Akerström S, Gunalan V, Keng CT, Tan YJ, Mirazimi A. Dual effect of nitric oxide on SARS-CoV replication: viral RNA production and palmitoylation of the S protein are affected. Virology 2009; 395:1-9. [PMID: 19800091 PMCID: PMC7111989 DOI: 10.1016/j.virol.2009.09.007] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Revised: 05/07/2009] [Accepted: 09/08/2009] [Indexed: 02/07/2023]
Abstract
Nitric oxide is an important molecule playing a key role in a broad range of biological process such as neurotransmission, vasodilatation and immune responses. While the anti-microbiological properties of nitric oxide-derived reactive nitrogen intermediates (RNI) such as peroxynitrite, are known, the mechanism of these effects are as yet poorly studied. Severe Acute Respiratory Syndrome coronavirus (SARS-CoV) belongs to the family Coronaviridae, was first identified during 2002-2003. Mortality in SARS patients ranges from between 6 to 55%. We have previously shown that nitric oxide inhibits the replication cycle of SARS-CoV in vitro by an unknown mechanism. In this study, we have further investigated the mechanism of the inhibition process of nitric oxide against SARS-CoV. We found that peroxynitrite, an intermediate product of nitric oxide in solution formed by the reaction of NO with superoxide, has no effect on the replication cycle of SARS-CoV, suggesting that the inhibition is either directly effected by NO or a derivative other than peroxynitrite. Most interestingly, we found that NO inhibits the replication of SARS-CoV by two distinct mechanisms. Firstly, NO or its derivatives cause a reduction in the palmitoylation of nascently expressed spike (S) protein which affects the fusion between the S protein and its cognate receptor, angiotensin converting enzyme 2. Secondly, NO or its derivatives cause a reduction in viral RNA production in the early steps of viral replication, and this could possibly be due to an effect on one or both of the cysteine proteases encoded in Orf1a of SARS-CoV.
Collapse
Affiliation(s)
- Sara Akerström
- Centre for Microbiological Preparedness, Swedish Institute for Infectious Disease Control, Nobels Väg 18, Solna, Sweden
| | | | | | | | | |
Collapse
|
45
|
Coughlin MM, Babcook J, Prabhakar BS. Human monoclonal antibodies to SARS-coronavirus inhibit infection by different mechanisms. Virology 2009; 394:39-46. [PMID: 19748648 PMCID: PMC7111986 DOI: 10.1016/j.virol.2009.07.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 06/22/2009] [Accepted: 07/15/2009] [Indexed: 12/28/2022]
Abstract
SARS-CoV causes an acute infection making targeted passive immunotherapy an attractive treatment strategy. We previously generated human mAbs specific to the S1 region of SARS-CoV S protein. These mAbs bind epitopes within the receptor binding domain (RBD) or upstream of the RBD. We show that mAbs recognizing epitopes within the RBD inhibit infection by preventing viral attachment to the cellular receptor. One mAb binds upstream of the RBD and prevents viral entry by inhibiting a post-binding event. Evaluation of several mAbs demonstrated varying ability of the mAbs to select escape mutants when used individually. However, a mixture of antibodies could effectively neutralize a range of mutant viruses. These data strongly suggest that a mixture containing antibodies recognizing distinct regions and targeting more than one step in viral entry is likely to be more effective in neutralizing the virus and suppressing the generation of escape mutants, and thus potentially constitute a highly effective passive immunotherapy.
Collapse
Affiliation(s)
- Melissa M Coughlin
- Department of Microbiology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | |
Collapse
|
46
|
Du L, He Y, Zhou Y, Liu S, Zheng BJ, Jiang S. The spike protein of SARS-CoV--a target for vaccine and therapeutic development. Nat Rev Microbiol 2009; 7:226-36. [PMID: 19198616 PMCID: PMC2750777 DOI: 10.1038/nrmicro2090] [Citation(s) in RCA: 1197] [Impact Index Per Article: 74.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This Review provides an overview on the spike (S) protein of severe acute respiratory syndrome-coronavirus (SARS-CoV) as a target for the development of vaccines and therapeutics for the prevention and treatment of SARS. SARS is a newly emerging infectious disease, caused by SARS-CoV, a novel coronavirus that caused a global outbreak of SARS. SARS-CoV S protein mediates binding of the virus with its receptor angiotensin-converting enzyme 2 and promotes the fusion between the viral and host cell membranes and virus entry into the host cell. SARS-CoV S protein induces humoral and cellular immune responses against SARS-CoV. SARS S protein is the target of new SARS vaccines. These vaccines are based on SARS-CoV full-length S protein and its receptor-binding domain, including DNA-, viral vector- and subunit-based vaccines Peptides, antibodies, organic compounds and short interfering RNAs are additional anti-SARS-CoV therapeutics that target the S protein. The work on SARS-CoV S protein-based vaccines and drugs will be useful as a model for the development of prophylactic strategies and therapies against other viruses with class I fusion proteins that can cause emerging infectious diseases.
The outbreaks of severe acute respiratory syndrome (SARS) between 2002 and 2004 killed hundreds of people. Vaccines against the SARS coronavirus (SARS-CoV) could protect the population during future outbreaks. In this Review, Shibo Jiang and colleagues describe such vaccines, as well as other therapeutics, based on the SARS-CoV spike protein. Severe acute respiratory syndrome (SARS) is a newly emerging infectious disease caused by a novel coronavirus, SARS-coronavirus (SARS-CoV). The SARS-CoV spike (S) protein is composed of two subunits; the S1 subunit contains a receptor-binding domain that engages with the host cell receptor angiotensin-converting enzyme 2 and the S2 subunit mediates fusion between the viral and host cell membranes. The S protein plays key parts in the induction of neutralizing-antibody and T-cell responses, as well as protective immunity, during infection with SARS-CoV. In this Review, we highlight recent advances in the development of vaccines and therapeutics based on the S protein.
Collapse
Affiliation(s)
- Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, 310 East 67th Street, New York, NY 10065, USA
| | | | | | | | | | | |
Collapse
|
47
|
Yan K, Tan W, Wang H, Wang Y, Zhang X, Li Y, Ruan L. SARS-CoV Spike Proteins Expressed by the Vaccinia Virus Tiantan Strain: Secreted SQ Protein Induces Robust Neutralization Antibody in Mice. Viral Immunol 2009; 22:57-66. [DOI: 10.1089/vim.2008.0064] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Kexia Yan
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Xuanwu District, Beijing, People's Republic of China
| | - Wenjie Tan
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Xuanwu District, Beijing, People's Republic of China
| | - Huijuan Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Xuanwu District, Beijing, People's Republic of China
| | - Yue Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Xuanwu District, Beijing, People's Republic of China
| | - Xiangmin Zhang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Xuanwu District, Beijing, People's Republic of China
| | - Yan Li
- National Microbiology Laboratory, Canadian Science Centre for Human and Animal Health, Winnipeg, Manitoba, Canada
| | - Li Ruan
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Xuanwu District, Beijing, People's Republic of China
| |
Collapse
|
48
|
Shen S, Mahadevappa G, Oh HLJ, Wee BY, Choi YW, Hwang LA, Lim SG, Hong W, Lal SK, Tan YJ. Comparing the antibody responses against recombinant hemagglutinin proteins of avian influenza A (H5N1) virus expressed in insect cells and bacteria. J Med Virol 2008; 80:1972-83. [PMID: 18814259 DOI: 10.1002/jmv.21298] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The hemagglutinin (HA) of influenza A virus plays an essential role in mediating the entry of the virus into host cells. Here, recombinant full-length HA5 protein from a H5N1 isolate (A/chicken/hatay/2004(H5N1)) was expressed and purified from the baculovirus-insect cell system. As expected, full-length HA5 elicits strong neutralizing antibodies, as evaluated in micro-neutralization tests using HA5 pseudotyped lentiviral particles. In addition, two fragments of HA5 were expressed in bacteria and the N-terminal fragment, covering the ectodomain before the HA1/HA2 polybasic cleavage site, was found to elicit neutralizing antibodies. But the C-terminal fragment, which covers the remaining portion of the ectodomain, did not. Neutralizing titer of the anti-serum against the N-terminal fragment is only four times lower than the anti-serum against the full-length HA5 protein. Using a novel membrane fusion assay, the abilities of these antibodies to block membrane fusion were found to correlate well with the neutralization activities.
Collapse
Affiliation(s)
- Shuo Shen
- Collaborative Anti-viral Research Group, Institute of Molecular and Cell Biology, Singapore, Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Suresh MR, Bhatnagar PK, Das D. Molecular targets for diagnostics and therapeutics of severe acute respiratory syndrome (SARS-CoV). JOURNAL OF PHARMACY AND PHARMACEUTICAL SCIENCES 2008; 11:1s-13s. [PMID: 19203466 DOI: 10.18433/j3j019] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE The large number of deaths in a short period of time due to the spread of severe acute respiratory syndrome (SARS) infection led to the unparalleled collaborative efforts world wide to determine and characterize the new coronavirus (SARS-CoV). The full genome sequence was determined within weeks of the first outbreak by the Canadian group with international collaboration. As per the World Health Organization (WHO), the continual lack of a rapid laboratory test to aid the early diagnosis of suspected cases of SARS makes this area a priority for future research. To prevent deaths in the future, early diagnosis and therapy of this infectious disease is of paramount importance. METHODS This review describes the specific molecular targets for diagnostics and therapeutics of viral infection. RESULTS The three major diagnostic methods available for SARS includes viral RNA detection by reverse transcription polymerase chain reaction (RT-PCR), virus induced antibodies by immunofluorescence assay (IFA) or by enzyme linked immunosorbant assay (ELISA) of nucleocapsid protein (NP). The spike glycoprotein of SARS-CoV is the major inducer of neutralizing antibodies. The receptor binding domain (RBD) in the S1 region of the spike glycoprotein contains multiple conformational epitopes that induces highly potent neutralizing antibodies. The genetically engineered attenuated form of the virus or viral vector vaccine encoding for the SARS-CoV spike glycoprotein has been shown to elicit protective immunity in vaccinated animals. CONCLUSION NP is the preferred target for routine detection of SARS-CoV infection by ELISA which is an economical method compared to other methods. The RBD of the spike glycoprotein is both a functional domain for cell receptor binding and also a major neutralizing determinant of SARS-CoV. The progress in evaluating a therapeutic or vaccine would depend on the avail ability of clinically relevant animal model.
Collapse
Affiliation(s)
- Mavanur R Suresh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| | | | | |
Collapse
|
50
|
Du L, Zhao G, Lin Y, Chan C, He Y, Jiang S, Wu C, Jin DY, Yuen KY, Zhou Y, Zheng BJ. Priming with rAAV encoding RBD of SARS-CoV S protein and boosting with RBD-specific peptides for T cell epitopes elevated humoral and cellular immune responses against SARS-CoV infection. Vaccine 2008; 26:1644-51. [PMID: 18289745 PMCID: PMC2600875 DOI: 10.1016/j.vaccine.2008.01.025] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Revised: 12/30/2007] [Accepted: 01/14/2008] [Indexed: 12/17/2022]
Abstract
Development of vaccines against severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV) is crucial in the prevention of SARS reemergence. The receptor-binding domain (RBD) of SARS-CoV spike (S) protein is an important target in developing safe and effective SARS vaccines. Our previous study has demonstrated that vaccination with adeno-associated virus encoding RBD (RBD-rAAV) induces high titer of neutralizing antibodies. In this study, we further assessed the immune responses and protective effect of the immunization with RBD-rAAV prime/RBD-specific T cell peptide boost. Compared with the RBD-rAAV prime/boost vaccination, RBD-rAAV prime/RBD-peptide (RBD-Pep) boost induced similar levels of Th1 and neutralizing antibody responses that protected the vaccinated mice from subsequent SARS-CoV challenge, but stronger Th2 and CTL responses. No significant immune responses and protective effects were detected in mice vaccinated with RBD-Pep or blank AAV alone. Since T cell epitopes are highly conserved and boosting with peptides may induce the production of effector memory T cells, which may be effective against viruses with mutations in the neutralizing epitopes, our results suggest that the vaccination protocol used may be ideal for providing effective, broad and long-term protection against SARS-CoV infection.
Collapse
Affiliation(s)
- Lanying Du
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|