301
|
Bauernfeind S, Salzberger B, Hitzenbichler F, Scigala K, Einhauser S, Wagner R, Gessner A, Koestler J, Peterhoff D. Association between Reactogenicity and Immunogenicity after Vaccination with BNT162b2. Vaccines (Basel) 2021; 9:1089. [PMID: 34696197 PMCID: PMC8538767 DOI: 10.3390/vaccines9101089] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 11/30/2022] Open
Abstract
It is not clear whether there is an association between adverse reactions and immune response after vaccination. Seven hundred and thirty-five vaccinees from our University Medical Center vaccination clinic provided information about sex, age and adverse reactions after first and second vaccination with BNT162b2. Adverse reactions were categorized into three groups: no or minor on the injection side, moderate (not further classified) and severe-defined as any symptom(s) resulting in sick leave. We chose 38 vaccinees with the most severe adverse reactions and compared their humoral and T-cell-mediated immune responses after second vaccination with those of 38 sex and age matched controls without or only minor injection-side related adverse reactions. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) anti-receptor binding domain (RBD) IgG titers were detectable in all participants (median 5528; range 958-26,285). Men with severe adverse reactions had 1.5-fold higher median SARS-CoV-2 RBD IgG titers compared to men without adverse reactions (median 7406 versus 4793; p < 0.001). Similarly; neutralization activity was significantly higher in men with severe adverse reactions (half maximal inhibitory concentrations (IC50) median 769 versus 485; p < 0.001). Reactogenicity did not influence humoral immune response in women nor T-cell-mediated immune response in any sex. To conclude; adverse reactions after vaccination with BNT162b2 do influence humoral immune response yet only in men and are not a prerequisite for a robust antibody response.
Collapse
Affiliation(s)
- Stilla Bauernfeind
- Department of Infection Prevention and Infectious Diseases, University Medical Center Regensburg, 93053 Regensburg, Germany; (B.S.); (F.H.); (K.S.)
| | - Bernd Salzberger
- Department of Infection Prevention and Infectious Diseases, University Medical Center Regensburg, 93053 Regensburg, Germany; (B.S.); (F.H.); (K.S.)
| | - Florian Hitzenbichler
- Department of Infection Prevention and Infectious Diseases, University Medical Center Regensburg, 93053 Regensburg, Germany; (B.S.); (F.H.); (K.S.)
| | - Karolina Scigala
- Department of Infection Prevention and Infectious Diseases, University Medical Center Regensburg, 93053 Regensburg, Germany; (B.S.); (F.H.); (K.S.)
| | - Sebastian Einhauser
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany; (S.E.); (R.W.); (A.G.); (D.P.)
| | - Ralf Wagner
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany; (S.E.); (R.W.); (A.G.); (D.P.)
- Institute for Clinical Microbiology and Hygiene, University Medical Center Regensburg, 93053 Regensburg, Germany;
| | - André Gessner
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany; (S.E.); (R.W.); (A.G.); (D.P.)
- Institute for Clinical Microbiology and Hygiene, University Medical Center Regensburg, 93053 Regensburg, Germany;
| | - Josef Koestler
- Institute for Clinical Microbiology and Hygiene, University Medical Center Regensburg, 93053 Regensburg, Germany;
| | - David Peterhoff
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany; (S.E.); (R.W.); (A.G.); (D.P.)
- Institute for Clinical Microbiology and Hygiene, University Medical Center Regensburg, 93053 Regensburg, Germany;
| |
Collapse
|
302
|
Law JLM, Logan M, Joyce MA, Landi A, Hockman D, Crawford K, Johnson J, LaChance G, Saffran HA, Shields J, Hobart E, Brassard R, Arutyunova E, Pabbaraju K, Croxen M, Tipples G, Lemieux MJ, Tyrrell DL, Houghton M. SARS-COV-2 recombinant Receptor-Binding-Domain (RBD) induces neutralizing antibodies against variant strains of SARS-CoV-2 and SARS-CoV-1. Vaccine 2021; 39:5769-5779. [PMID: 34481699 PMCID: PMC8387217 DOI: 10.1016/j.vaccine.2021.08.081] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 12/11/2022]
Abstract
SARS-CoV-2 is the etiological agent of COVID19. There are currently several licensed vaccines approved for human use and most of them target the spike protein in the virion envelope to induce protective immunity. Recently, variants that spread more quickly have emerged. There is evidence that some of these variants are less sensitive to neutralization in vitro, but it is not clear whether they can evade vaccine induced protection. In this study, we tested SARS-CoV-2 spike RBD as a vaccine antigen and explored the effect of formulation with Alum/MPLA or AddaS03 adjuvants. Our results show that RBD induces high titers of neutralizing antibodies and activates strong cellular immune responses. There is also significant cross-neutralization of variants B.1.1.7 and B.1.351 and to a lesser extent, SARS-CoV-1. These results indicate that recombinant RBD can be a viable candidate as a stand-alone vaccine or as a booster shot to diversify our strategy for COVID19 protection.
Collapse
Affiliation(s)
- John Lok Man Law
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada.
| | - Michael Logan
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Michael A Joyce
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Abdolamir Landi
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Darren Hockman
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Kevin Crawford
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Janelle Johnson
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Gerald LaChance
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Holly A Saffran
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Justin Shields
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Eve Hobart
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Raelynn Brassard
- Department of Biochemistry, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Elena Arutyunova
- Department of Biochemistry, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | | | | | - Graham Tipples
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada; Alberta Precision Laboratories, Edmonton, Canada
| | - M Joanne Lemieux
- Department of Biochemistry, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - D Lorne Tyrrell
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Michael Houghton
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada.
| |
Collapse
|
303
|
Rosati M, Agarwal M, Hu X, Devasundaram S, Stellas D, Chowdhury B, Bear J, Burns R, Donohue D, Pessaint L, Andersen H, Lewis MG, Terpos E, Dimopoulos MA, Wlodawer A, Mullins JI, Venzon DJ, Pavlakis GN, Felber BK. Control of SARS-CoV-2 infection after Spike DNA or Spike DNA+Protein co-immunization in rhesus macaques. PLoS Pathog 2021; 17:e1009701. [PMID: 34551020 PMCID: PMC8489704 DOI: 10.1371/journal.ppat.1009701] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/04/2021] [Accepted: 09/07/2021] [Indexed: 12/28/2022] Open
Abstract
The speed of development, versatility and efficacy of mRNA-based vaccines have been amply demonstrated in the case of SARS-CoV-2. DNA vaccines represent an important alternative since they induce both humoral and cellular immune responses in animal models and in human trials. We tested the immunogenicity and protective efficacy of DNA-based vaccine regimens expressing different prefusion-stabilized Wuhan-Hu-1 SARS-CoV-2 Spike antigens upon intramuscular injection followed by electroporation in rhesus macaques. Different Spike DNA vaccine regimens induced antibodies that potently neutralized SARS-CoV-2 in vitro and elicited robust T cell responses. The antibodies recognized and potently neutralized a panel of different Spike variants including Alpha, Delta, Epsilon, Eta and A.23.1, but to a lesser extent Beta and Gamma. The DNA-only vaccine regimens were compared to a regimen that included co-immunization of Spike DNA and protein in the same anatomical site, the latter of which showed significant higher antibody responses. All vaccine regimens led to control of SARS-CoV-2 intranasal/intratracheal challenge and absence of virus dissemination to the lower respiratory tract. Vaccine-induced binding and neutralizing antibody titers and antibody-dependent cellular phagocytosis inversely correlated with transient virus levels in the nasal mucosa. Importantly, the Spike DNA+Protein co-immunization regimen induced the highest binding and neutralizing antibodies and showed the strongest control against SARS-CoV-2 challenge in rhesus macaques. Anti-Spike neutralizing antibodies provide strong protection against SARS-CoV-2 infection in animal models, and correlate with protection in humans, supporting the notion that induction of strong humoral immunity is key to protection. We show induction of robust antibody and T cell responses by different Spike DNA-based vaccine regimens able to effectively mediate protection and to control SARS-CoV-2 infection in the rhesus macaque model. This study provides the opportunity to compare vaccines able to induce different humoral and cellular immune responses in an effort to develop durable immunity against the SARS-CoV-2. A vaccine regimen comprising simultaneous co-immunization of DNA and Protein at the same anatomical site showed best neutralizing abilities and was more effective than DNA alone in inducing protective immune responses and controlling SARS-CoV-2 infection. Thus, an expansion of the DNA vaccine regimen to include co-immunization with Spike protein may be of advantage also for SARS-CoV-2.
Collapse
Affiliation(s)
- Margherita Rosati
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Mahesh Agarwal
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Xintao Hu
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Santhi Devasundaram
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Dimitris Stellas
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Bhabadeb Chowdhury
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Robert Burns
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Duncan Donohue
- MS Applied Information and Management Sciences, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | | | - Hanne Andersen
- BIOQUAL, Inc.; Rockville, Maryland, United States of America
| | - Mark G. Lewis
- BIOQUAL, Inc.; Rockville, Maryland, United States of America
| | - Evangelos Terpos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | | | - Alexander Wlodawer
- Center for Structural Biology, National Cancer Institute, Frederick, Maryland, United States of America
| | - James I. Mullins
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - David J. Venzon
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
304
|
Molnar T, Varnai R, Schranz D, Zavori L, Peterfi Z, Sipos D, Tőkés-Füzesi M, Illes Z, Buki A, Csecsei P. Severe Fatigue and Memory Impairment Are Associated with Lower Serum Level of Anti-SARS-CoV-2 Antibodies in Patients with Post-COVID Symptoms. J Clin Med 2021; 10:jcm10194337. [PMID: 34640355 PMCID: PMC8509483 DOI: 10.3390/jcm10194337] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Post-COVID manifestation is defined as persistent symptoms or long-term complications beyond 4 weeks from disease onset. Fatigue and memory impairment are common post-COVID symptoms. We aimed to explore associations between the timeline and severity of post-COVID fatigue and anti-SARS-CoV-2 antibodies. Methods: Fatigue and memory impairment were assessed in a total of 101 post-COVID subjects using the Chalder fatigue scale (CFQ-11) and a visual analogue scale. Using the bimodal scoring system generated from CFQ-11, a score ≥4 was defined as severe fatigue. Serum anti-SARS-CoV-2 spike (anti-S-Ig) and nucleocapsid (anti-NC-Ig) antibodies were examined at two time points: 4–12 weeks after onset of symptoms, and beyond 12 weeks. Results: The serum level of anti-S-Ig was significantly higher in patients with non-severe fatigue compared to those with severe fatigue at 4–12 weeks (p = 0.006) and beyond 12 weeks (p = 0.016). The serum level of anti-NC-Ig remained high in patients with non-severe fatigue at both time points. In contrast, anti-NC-Ig decreased significantly in severe fatigue cases regardless of the elapsed time (4–12 weeks: p = 0.024; beyond 12 weeks: p = 0.005). The incidence of memory impairment was significantly correlated with lower anti-S-Ig levels (−0.359, p < 0.001). Conclusion: The systemic immune response reflected by antibodies to SARS-CoV-2 is strongly correlated with the severity of post-COVID fatigue.
Collapse
Affiliation(s)
- Tihamer Molnar
- Department of Anaesthesiology and Intensive Care, University of Pecs, Medical School, H7632 Pecs, Hungary;
| | - Reka Varnai
- Department of Primary Health Care, University of Pecs, Medical School, H7632 Pecs, Hungary
- Correspondence: ; Tel.: +36-72535900
| | - Daniel Schranz
- Department of Neurology, University of Pecs, Medical School, H7632 Pecs, Hungary;
| | - Laszlo Zavori
- Salisbury NHS Foundation Trust, Salisbury SP2 8BJ, UK;
| | - Zoltan Peterfi
- 1st Department of Internal Medicine, Division of Infectology, University of Pecs, Medical School, H7632 Pecs, Hungary; (Z.P.); (D.S.)
| | - David Sipos
- 1st Department of Internal Medicine, Division of Infectology, University of Pecs, Medical School, H7632 Pecs, Hungary; (Z.P.); (D.S.)
| | - Margit Tőkés-Füzesi
- Department of Laboratory Medicine, University of Pecs, Medical School, H7632 Pecs, Hungary;
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, University of Southern Denmark, 5230 Odense, Denmark;
| | - Andras Buki
- Department of Neurosurgery, University of Pecs, Medical School, H7632 Pecs, Hungary; (A.B.); (P.C.)
| | - Peter Csecsei
- Department of Neurosurgery, University of Pecs, Medical School, H7632 Pecs, Hungary; (A.B.); (P.C.)
| |
Collapse
|
305
|
Sholukh AM, Fiore-Gartland A, Ford ES, Miner MD, Hou YJ, Tse LV, Kaiser H, Zhu H, Lu J, Madarampalli B, Park A, Lempp FA, St. Germain R, Bossard EL, Kee JJ, Diem K, Stuart AB, Rupert PB, Brock C, Buerger M, Doll MK, Randhawa AK, Stamatatos L, Strong RK, McLaughlin C, Huang ML, Jerome KR, Baric RS, Montefiori D, Corey L. Evaluation of Cell-Based and Surrogate SARS-CoV-2 Neutralization Assays. J Clin Microbiol 2021; 59:e0052721. [PMID: 34288726 PMCID: PMC8451402 DOI: 10.1128/jcm.00527-21] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/25/2021] [Indexed: 11/22/2022] Open
Abstract
Determinants of protective immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection require the development of well-standardized, reproducible antibody assays. This need has led to the emergence of a variety of neutralization assays. Head-to-head evaluation of different SARS-CoV-2 neutralization platforms could facilitate comparisons across studies and laboratories. Five neutralization assays were compared using 40 plasma samples from convalescent individuals with mild to moderate coronavirus disease 2019 (COVID-19): four cell-based systems using either live recombinant SARS-CoV-2 or pseudotyped viral particles created with lentivirus (LV) or vesicular stomatitis virus (VSV) packaging and one surrogate enzyme-linked immunosorbent assay (ELISA)-based test that measures inhibition of the spike protein receptor binding domain (RBD) binding its receptor human angiotensin converting enzyme 2 (hACE2). Vero cells, Vero E6 cells, HEK293T cells expressing hACE2, and TZM-bl cells expressing hACE2 and transmembrane serine protease 2 were tested. All cell-based assays showed 50% neutralizing dilution (ND50) geometric mean titers (GMTs) that were highly correlated (Pearson r = 0.81 to 0.89) and ranged within 3.4-fold. The live virus assay and LV pseudovirus assays with HEK293T/hACE2 cells showed very similar mean titers, 141 and 178, respectively. ND50 titers positively correlated with plasma IgG targeting SARS-CoV-2 spike protein and RBD (r = 0.63 to 0.89), but moderately correlated with nucleoprotein IgG (r = 0.46 to 0.73). ND80 GMTs mirrored ND50 data and showed similar correlation between assays and with IgG concentrations. The VSV pseudovirus assay and LV pseudovirus assay with HEK293T/hACE2 cells in low- and high-throughput versions were calibrated against the WHO SARS-CoV-2 IgG standard. High concordance between the outcomes of cell-based assays with live and pseudotyped virions enables valid cross-study comparison using these platforms.
Collapse
Affiliation(s)
- Anton M. Sholukh
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
| | - Andrew Fiore-Gartland
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
| | - Emily S. Ford
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Maurine D. Miner
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
| | - Yixuan J. Hou
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Longping V. Tse
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Haiying Zhu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Joyce Lu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Bhanupriya Madarampalli
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Arnold Park
- Vir Biotechnology, San Francisco, California, USA
| | | | - Russell St. Germain
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
| | - Emily L. Bossard
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
| | - Jia Jin Kee
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
| | - Kurt Diem
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Andrew B. Stuart
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
| | - Peter B. Rupert
- Basic Sciences Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
| | - Chance Brock
- Basic Sciences Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
| | - Matthew Buerger
- Basic Sciences Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
| | - Margaret K. Doll
- Department of Population Health Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York, USA
| | - April Kaur Randhawa
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
| | - Leonidas Stamatatos
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
| | - Roland K. Strong
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
- Basic Sciences Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
| | - Colleen McLaughlin
- Department of Population Health Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York, USA
| | - Meei-Li Huang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Keith R. Jerome
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - David Montefiori
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Lawrence Corey
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, Washington, USA
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
306
|
Kinetics of Nucleocapsid, Spike and Neutralizing Antibodies, and Viral Load in Patients with Severe COVID-19 Treated with Convalescent Plasma. Viruses 2021; 13:v13091844. [PMID: 34578426 PMCID: PMC8473255 DOI: 10.3390/v13091844] [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: 07/17/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/16/2022] Open
Abstract
COVID-19 is an ongoing pandemic with high morbidity and mortality. Despite meticulous research, only dexamethasone has shown consistent mortality reduction. Convalescent plasma (CP) infusion might also develop into a safe and effective treatment modality on the basis of recent studies and meta-analyses; however, little is known regarding the kinetics of antibodies in CP recipients. To evaluate the kinetics, we followed 31 CP recipients longitudinally enrolled at a median of 3 days post symptom onset for changes in binding and neutralizing antibody titers and viral loads. Antibodies against the complete trimeric Spike protein and the receptor-binding domain (Spike-RBD), as well as against the complete Nucleocapsid protein and the RNA binding domain (N-RBD) were determined at baseline and weekly following CP infusion. Neutralizing antibody (pseudotype NAb) titers were determined at the same time points. Viral loads were determined semi-quantitatively by SARS-CoV-2 PCR. Patients with low humoral responses at entry showed a robust increase of antibodies to all SARS-CoV-2 proteins and Nab, reaching peak levels within 2 weeks. The rapid increase in binding and neutralizing antibodies was paralleled by a concomitant clearance of the virus within the same timeframe. Patients with high humoral responses at entry demonstrated low or no further increases; however, virus clearance followed the same trajectory as in patients with low antibody response at baseline. Together, the sequential immunological and virological analysis of this well-defined cohort of patients early in infection shows the presence of high levels of binding and neutralizing antibodies and potent clearance of the virus.
Collapse
|
307
|
Caniels TG, Bontjer I, van der Straten K, Poniman M, Burger JA, Appelman B, Lavell HAA, Oomen M, Godeke GJ, Valle C, Mögling R, van Willigen HDG, Wynberg E, Schinkel M, van Vught LA, Guerra D, Snitselaar JL, Chaturbhuj DN, Cuella Martin I, Amsterdam UMC COVID-19 S3/HCW study group, Moore JP, de Jong MD, Reusken C, Sikkens JJ, Bomers MK, de Bree GJ, van Gils MJ, Eggink D, Sanders RW. Emerging SARS-CoV-2 variants of concern evade humoral immune responses from infection and vaccination. SCIENCE ADVANCES 2021; 7:eabj5365. [PMID: 34516917 PMCID: PMC8442901 DOI: 10.1126/sciadv.abj5365] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/13/2021] [Indexed: 05/21/2023]
Abstract
Emerging SARS-CoV-2 variants of concern (VOCs) pose a threat to human immunity induced by natural infection and vaccination. We assessed the recognition of three VOCs (B.1.1.7, B.1.351, and P.1) in cohorts of COVID-19 convalescent patients (n = 69) and Pfizer-BioNTech vaccine recipients (n = 50). Spike binding and neutralization against all three VOCs were substantially reduced in most individuals, with the largest four- to sevenfold reduction in neutralization being observed against B.1.351. While hospitalized patients with COVID-19 and vaccinees maintained sufficient neutralizing titers against all three VOCs, 39% of nonhospitalized patients exhibited no detectable neutralization against B.1.351. Moreover, monoclonal neutralizing antibodies show sharp reductions in their binding kinetics and neutralizing potential to B.1.351 and P.1 but not to B.1.1.7. These data have implications for the degree to which pre-existing immunity can protect against subsequent infection with VOCs and informs policy makers of susceptibility to globally circulating SARS-CoV-2 VOCs.
Collapse
Affiliation(s)
- Tom G. Caniels
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Ilja Bontjer
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Karlijn van der Straten
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Meliawati Poniman
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Judith A. Burger
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Brent Appelman
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - H. A. Ayesha Lavell
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Melissa Oomen
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Gert-Jan Godeke
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Coralie Valle
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Ramona Mögling
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Hugo D. G. van Willigen
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Elke Wynberg
- Department of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Public Health Service of Amsterdam, Amsterdam, Netherlands
| | - Michiel Schinkel
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Lonneke A. van Vught
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Denise Guerra
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Jonne L. Snitselaar
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Devidas N. Chaturbhuj
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - Isabel Cuella Martin
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Amsterdam UMC COVID-19 S3/HCW study group
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
- Department of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Public Health Service of Amsterdam, Amsterdam, Netherlands
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - John P. Moore
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - Menno D. de Jong
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Chantal Reusken
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Jonne J. Sikkens
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Marije K. Bomers
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Godelieve J. de Bree
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Marit J. van Gils
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Dirk Eggink
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Rogier W. Sanders
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
308
|
Mishra N, Kumar S, Singh S, Bansal T, Jain N, Saluja S, Kumar R, Bhattacharyya S, Palanichamy JK, Mir RA, Sinha S, Luthra K. Cross-neutralization of SARS-CoV-2 by HIV-1 specific broadly neutralizing antibodies and polyclonal plasma. PLoS Pathog 2021; 17:e1009958. [PMID: 34559854 PMCID: PMC8494312 DOI: 10.1371/journal.ppat.1009958] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 10/06/2021] [Accepted: 09/15/2021] [Indexed: 11/30/2022] Open
Abstract
Cross-reactive epitopes (CREs) are similar epitopes on viruses that are recognized or neutralized by same antibodies. The S protein of SARS-CoV-2, similar to type I fusion proteins of viruses such as HIV-1 envelope (Env) and influenza hemagglutinin, is heavily glycosylated. Viral Env glycans, though host derived, are distinctly processed and thereby recognized or accommodated during antibody responses. In recent years, highly potent and/or broadly neutralizing human monoclonal antibodies (bnAbs) that are generated in chronic HIV-1 infections have been defined. These bnAbs exhibit atypical features such as extensive somatic hypermutations, long complementary determining region (CDR) lengths, tyrosine sulfation and presence of insertions/deletions, enabling them to effectively neutralize diverse HIV-1 viruses despite extensive variations within the core epitopes they recognize. As some of the HIV-1 bnAbs have evolved to recognize the dense viral glycans and cross-reactive epitopes (CREs), we assessed if these bnAbs cross-react with SARS-CoV-2. Several HIV-1 bnAbs showed cross-reactivity with SARS-CoV-2 while one HIV-1 CD4 binding site bnAb, N6, neutralized SARS-CoV-2. Furthermore, neutralizing plasma antibodies of chronically HIV-1 infected children showed cross neutralizing activity against SARS-CoV-2 pseudoviruses. Collectively, our observations suggest that human monoclonal antibodies tolerating extensive epitope variability can be leveraged to neutralize pathogens with related antigenic profile.
Collapse
Affiliation(s)
- Nitesh Mishra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sanjeev Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
- ICGEB-Emory Vaccine Centre Program, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Swarandeep Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Tanu Bansal
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Nishkarsh Jain
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sumedha Saluja
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Rajesh Kumar
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Sankar Bhattacharyya
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | | | - Riyaz Ahmad Mir
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subrata Sinha
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
309
|
Van Rompay KKA, Olstad KJ, Sammak RL, Dutra J, Watanabe JK, Usachenko JL, Immareddy R, Roh JW, Verma A, Shaan Lakshmanappa Y, Schmidt BA, Di Germanio C, Rizvi N, Stone M, Simmons G, Dumont LJ, Allen AM, Lockwood S, Pollard RE, de Assis RR, Yee JL, Nham PB, Ardeshir A, Deere JD, Patterson J, Jain A, Felgner PL, Iyer SS, Hartigan-O'Connor DJ, Busch MP, Reader JR. Early post-infection treatment of SARS-CoV-2 infected macaques with human convalescent plasma with high neutralizing activity reduces lung inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.09.01.458520. [PMID: 34494025 DOI: 10.1101/2021.08.06.455491] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
UNLABELLED Early in the SARS-CoV-2 pandemic, there was a high level of optimism based on observational studies and small controlled trials that treating hospitalized patients with convalescent plasma from COVID-19 survivors (CCP) would be an important immunotherapy. However, as more data from controlled trials became available, the results became disappointing, with at best moderate evidence of efficacy when CCP with high titers of neutralizing antibodies was used early in infection. To better understand the potential therapeutic efficacy of CCP, and to further validate SARS-CoV-2 infection of macaques as a reliable animal model for testing such strategies, we inoculated 12 adult rhesus macaques with SARS-CoV-2 by intratracheal and intranasal routes. One day later, 8 animals were infused with pooled human CCP with a high titer of neutralizing antibodies (RVPN NT 50 value of 3,003), while 4 control animals received normal human plasma. Animals were monitored for 7 days. Animals treated with CCP had detectable levels of antiviral antibodies after infusion. In comparison to the control animals, they had similar levels of virus replication in the upper and lower respiratory tract, but had significantly reduced interstitial pneumonia, as measured by comprehensive lung histology. By highlighting strengths and weaknesses, data of this study can help to further optimize nonhuman primate models to provide proof-of-concept of intervention strategies, and guide the future use of convalescent plasma against SARS-CoV-2 and potentially other newly emerging respiratory viruses. AUTHOR SUMMARY The results of treating SARS-CoV-2 infected hospitalized patients with COVID-19 convalescent plasma (CCP), collected from survivors of natural infection, have been disappointing. The available data from various studies indicate at best moderate clinical benefits only when CCP with high titer of neutralizing antibodies was infused early in infection. The macaque model of SARS-CoV-2 infection can be useful to gain further insights in the value of CCP therapy. In this study, animals were infected with SARS-CoV-2 and the next day, were infused with pooled human convalescent plasma, selected to have a very high titer of neutralizing antibodies. While administration of CCP did not result in a detectable reduction in virus replication in the respiratory tract, it significantly reduced lung inflammation. These data, combined with the results of monoclonal antibody studies, emphasize the need to use products with high titers of neutralizing antibodies, and guide the future development of CCP-based therapies.
Collapse
Affiliation(s)
- Koen K A Van Rompay
- California National Primate Research Center, University of California, Davis, CA 95616
- Department of Pathology, Microbiology and Immunology, University of California, Davis, CA 95616
| | - Katherine J Olstad
- California National Primate Research Center, University of California, Davis, CA 95616
- Department of Pathology, Microbiology and Immunology, University of California, Davis, CA 95616
| | - Rebecca L Sammak
- California National Primate Research Center, University of California, Davis, CA 95616
| | - Joseph Dutra
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA 95616
| | - Jennifer K Watanabe
- California National Primate Research Center, University of California, Davis, CA 95616
| | - Jodie L Usachenko
- California National Primate Research Center, University of California, Davis, CA 95616
| | - Ramya Immareddy
- California National Primate Research Center, University of California, Davis, CA 95616
| | - Jamin W Roh
- Center for Immunology and Infectious Diseases, University of California, Davis, CA 95616
- Graduate Group in Immunology, University of California, Davis, CA 95616
| | - Anil Verma
- Center for Immunology and Infectious Diseases, University of California, Davis, CA 95616
| | | | - Brian A Schmidt
- Center for Immunology and Infectious Diseases, University of California, Davis, CA 95616
| | | | - Nabeela Rizvi
- Vitalant Research Institute, San Francisco, CA 94118
| | - Mars Stone
- Vitalant Research Institute, San Francisco, CA 94118
| | | | - Larry J Dumont
- Vitalant Research Institute, Denver, CO 80230; University of Colorado School of Medicine, Aurora, CO 80045
| | - A Mark Allen
- California National Primate Research Center, University of California, Davis, CA 95616
| | - Sarah Lockwood
- California National Primate Research Center, University of California, Davis, CA 95616
| | - Rachel E Pollard
- School of Veterinary Medicine, University of California, Davis, CA 95616
| | - Rafael Ramiro de Assis
- Vaccine Research and Development Center, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA 92697
| | - JoAnn L Yee
- California National Primate Research Center, University of California, Davis, CA 95616
| | - Peter B Nham
- California National Primate Research Center, University of California, Davis, CA 95616
| | - Amir Ardeshir
- California National Primate Research Center, University of California, Davis, CA 95616
| | - Jesse D Deere
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA 95616
| | - Jean Patterson
- Translational Research Section, Virology Branch, DMID/NIAID/NIH, MD 20852
| | - Aarti Jain
- Vaccine Research and Development Center, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA 92697
| | - Philip L Felgner
- Vaccine Research and Development Center, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA 92697
| | - Smita S Iyer
- California National Primate Research Center, University of California, Davis, CA 95616
- Department of Pathology, Microbiology and Immunology, University of California, Davis, CA 95616
- Center for Immunology and Infectious Diseases, University of California, Davis, CA 95616
| | - Dennis J Hartigan-O'Connor
- California National Primate Research Center, University of California, Davis, CA 95616
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA 95616
| | - Michael P Busch
- Vitalant Research Institute, San Francisco, CA 94118
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94118
| | - J Rachel Reader
- California National Primate Research Center, University of California, Davis, CA 95616
- Department of Pathology, Microbiology and Immunology, University of California, Davis, CA 95616
| |
Collapse
|
310
|
McDonald L, Wise H, Muecksch F, Poston D, Mavin S, Templeton K, Furrie E, Richardson C, McGuire J, Jarvis L, Malloy K, McAuley A, Palmateer N, Dickson E, Hatziioannou T, Bieniasz P, Jenks S. Comparison of SARS-CoV-2 serological assays for use in epidemiological surveillance in Scotland. JOURNAL OF CLINICAL VIROLOGY PLUS 2021; 1:100028. [PMID: 35262014 PMCID: PMC8200328 DOI: 10.1016/j.jcvp.2021.100028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 11/18/2022] Open
Abstract
Background Sero-surveillance of SARS-CoV-2 is crucial to monitoring levels of population exposure and informing public health responses, but may be influenced by variability in performance between available assays. Methods Five commercial immunoassays and a neutralising activity assay were used to detect antibodies to SARS-CoV-2 in routine primary care and paediatric samples collected during the first wave of the pandemic in NHS Lothian, Scotland as part of ongoing surveillance efforts. For each assay, sensitivity and specificity was calculated relative to consensus results (majority of immunoassays positive = overall positive) and neutralising activity. Quantitative correlation was performed between serological and neutralising titres. Results Seroprevalence ranged from 3.4-7.3 % in primary care patients and 3-5.9 % in paediatric patients according to different immunoassays. Neutralising activity was detectable in 2.8 % and 1.3 % respectively. Relative assay performance changed depending on comparison to immunoassay consensus versus neutralising activity and qualititative versus quantitative agreement. Cross-reactivity with endemic seasonal coronaviruses was confirmed by neutralising assay in false positives for one immunoassay. Presence of false positives for another assay was found specifically in paediatric but not adult samples. Conclusions Five serological assays show variable accuracy when applied to the general population, impacting seroprevalence estimates. Assay performance may also vary in detection of protective neutralising antibody levels. These aspects should be considered in assay selection and interpretation in epidemiological studies.
Collapse
Affiliation(s)
| | - Helen Wise
- Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, Scotland
| | - Frauke Muecksch
- Laboratory of Retrovirology, The Rockefeller University, New York, New York, USA
| | - Daniel Poston
- Laboratory of Retrovirology, The Rockefeller University, New York, New York, USA
| | - Sally Mavin
- Scottish Microbiology Reference Laboratory, NHS Highland, Inverness, Scotland
| | - Kate Templeton
- Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, Scotland
| | - Elizabeth Furrie
- Ninewells Hospital and Medical School, NHS Tayside, Dundee, Scotland
| | | | | | - Lisa Jarvis
- Scottish National Blood Transfusion Service, The Jack Copland Centre, Edinburgh, Scotland
| | - Kristen Malloy
- Scottish National Blood Transfusion Service, The Jack Copland Centre, Edinburgh, Scotland
| | - Andrew McAuley
- Public Health Scotland, Glasgow, Scotland
- School of Health & Life Sciences, Glasgow Caledonian University, Glasgow, Scotland
| | - Norah Palmateer
- Public Health Scotland, Glasgow, Scotland
- School of Health & Life Sciences, Glasgow Caledonian University, Glasgow, Scotland
| | | | | | - Paul Bieniasz
- Laboratory of Retrovirology, The Rockefeller University, New York, New York, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, New York, USA
| | - Sara Jenks
- Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, Scotland
| |
Collapse
|
311
|
Hasenkrug KJ, Feldmann F, Myers L, Santiago ML, Guo K, Barrett BS, Mickens KL, Carmody A, Okumura A, Rao D, Collins MM, Messer RJ, Lovaglio J, Shaia C, Rosenke R, van Doremalen N, Clancy C, Saturday G, Hanley P, Smith BJ, Meade-White K, Shupert WL, Hawman DW, Feldmann H. Recovery from Acute SARS-CoV-2 Infection and Development of Anamnestic Immune Responses in T Cell-Depleted Rhesus Macaques. mBio 2021; 12:e0150321. [PMID: 34311582 PMCID: PMC8406331 DOI: 10.1128/mbio.01503-21] [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] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 06/21/2021] [Indexed: 12/16/2022] Open
Abstract
Severe coronavirus disease 2019 (COVID-19) has been associated with T cell lymphopenia, but no causal effect of T cell deficiency on disease severity has been established. To investigate the specific role of T cells in recovery from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, we studied rhesus macaques that were depleted of either CD4+, CD8+, or both T cell subsets prior to infection. Peak virus loads were similar in all groups, but the resolution of virus in the T cell-depleted animals was slightly delayed compared to that in controls. The T cell-depleted groups developed virus-neutralizing antibody responses and class switched to IgG. When reinfected 6 weeks later, the T cell-depleted animals showed anamnestic immune responses characterized by rapid induction of high-titer virus-neutralizing antibodies, faster control of virus loads, and reduced clinical signs. These results indicate that while T cells play a role in the recovery of rhesus macaques from acute SARS-CoV-2 infections, their depletion does not induce severe disease, and T cells do not account for the natural resistance of rhesus macaques to severe COVID-19. Neither primed CD4+ nor CD8+ T cells appeared critical for immunoglobulin class switching, the development of immunological memory, or protection from a second infection. IMPORTANCE Patients with severe COVID-19 often have decreased numbers of T cells, a cell type important in fighting most viral infections. However, it is not known whether the loss of T cells contributes to severe COVID-19 or is a consequence of it. We studied rhesus macaques, which develop only mild COVID-19, similar to most humans. Experimental depletion of T cells slightly prolonged their clearance of virus, but there was no increase in disease severity. Furthermore, they were able to develop protection from a second infection and produced antibodies capable of neutralizing the virus. They also developed immunological memory, which allows a much stronger and more rapid response upon a second infection. These results suggest that T cells are not critical for recovery from acute SARS-CoV-2 infections in this model and point toward B cell responses and antibodies as the essential mediators of protection from re-exposure.
Collapse
Affiliation(s)
- Kim J. Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Lara Myers
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Mario L. Santiago
- Departments of Medicine, Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kejun Guo
- Departments of Medicine, Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Bradley S. Barrett
- Departments of Medicine, Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kaylee L. Mickens
- Departments of Medicine, Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Aaron Carmody
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Atsushi Okumura
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Deepashri Rao
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Madison M. Collins
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Ronald J. Messer
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Rebecca Rosenke
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Neeltje van Doremalen
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Chad Clancy
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Patrick Hanley
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Brian J. Smith
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Kimberly Meade-White
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - W. Lesley Shupert
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - David W. Hawman
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Heinz Feldmann
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| |
Collapse
|
312
|
Shang Y, Chen F, Li S, Song L, Gao Y, Yu X, Zheng J. Investigation of Interaction between the Spike Protein of SARS-CoV-2 and ACE2-Expressing Cells Using an In Vitro Cell Capturing System. Biol Proced Online 2021; 23:16. [PMID: 34433426 PMCID: PMC8387204 DOI: 10.1186/s12575-021-00153-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/27/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The Interaction between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein with Angiotensin converting enzyme 2 (ACE2) on the host cells is a crucial step for the viral entry and infection. Therefore, investigating the molecular mechanism underlying the interaction is of great importance for the prevention of the infection of SARS-CoV-2. In this study, we aimed to establish a virus-free in vitro system to study the interaction between the spike protein and host cells of SARS-CoV-2. RESULTS Our results show that ACE2-overexpressing HEK293T cells are captured by immobilized spike S1 protein, and the cell capturing process can be inhibited by the receptor binding domain of the spike protein or antibodies against S protein. Furthermore, spike S1 protein variant with D614G mutant show a higher cell capturing ability than wild type spike S1 protein and stronger binding capacity of its receptor ACE2. In addition, the captured cells can be eluted as living cells for further investigation. CONCLUSIONS This study provides a new in vitro system for investigating the interaction between SARS-CoV-2 and host cells and purifying ACE2-expressing cells.
Collapse
Affiliation(s)
- Yuning Shang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, XinXiang, China
| | - Feixiang Chen
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, XinXiang, China
| | - Shasha Li
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, XinXiang, China
| | - Lijuan Song
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, XinXiang, China
| | - Yunzhen Gao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, XinXiang, China
| | - Xinhua Yu
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), Borstel, Germany.
| | - Junfeng Zheng
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, XinXiang, China.
| |
Collapse
|
313
|
He X, Ding L, Cao K, Peng H, Gu C, Li Y, Li D, Dong L, Hong X, Wang X, Fu M, Qiu C, Zhu C, Zhang Z, Song S, Wang C, Jiang Z, Xie Y, Qi Z, Zhao C, Zhao P, Zhang X, Xu J. A human cell-based SARS-CoV-2 vaccine elicits potent neutralizing antibody responses and protects mice from SARS-CoV-2 challenge. Emerg Microbes Infect 2021; 10:1555-1573. [PMID: 34304724 PMCID: PMC8366622 DOI: 10.1080/22221751.2021.1957400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
To curb the pandemic of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), multiple platforms have been employed toward a safe and highly effective vaccine. Here, we develop a novel cell-based vaccine candidate, namely K562-S, by utilizing human cell K562 as a cellular carrier to display Spike (S) protein of SARS-CoV-2 on the membrane. Analogous to the traditional inactivated vaccine, K562-S cells can be propagated to a large scale by culturing and completely lose their viability after exposure to X-ray irradiation or formalin. We in turn demonstrated high immunogenicity of formalin-inactivated K562-S vaccine in both mouse and non-human primates and its protective efficacy in mice. In mice, immunization with inactivated K562-S vaccines can elicit potent neutralizing antibody (nAb) responses persisting longer than 5 months. We consequently showed in a hACE2 mouse model of SARS-CoV-2 infection that a two-shot vaccination with adjuvanted K562-S rendered greater than 3 log reduction in viral lung load and concomitant ameliorated lung pathology. Of importance, the administration of the same regimen in non-human primates was able to induce a neutralizing antibody titer averaging three-fold higher relative to human convalescent serum. These results together support the promise of K562-based, S-protein-expressing vaccines as a novel vaccination approach against SARS-CoV-2. Importantly, with a powerful capacity to carry external genes for cell-based vectors, this platform could rapidly generate two- and multiple-valent vaccines by incorporating SARS-CoV-2 mutants, SARS-CoV, or MERS-CoV.
Collapse
Affiliation(s)
- Xiangchuan He
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Longfei Ding
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Kangli Cao
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Haoran Peng
- Department of Microbiology, Second Military Medical University, Shanghai, People's Republic of China
| | - Chenjian Gu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yutang Li
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Duoduo Li
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Lanlan Dong
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Xiujing Hong
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Xiangwei Wang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Meilan Fu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Chenli Qiu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Cuisong Zhu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Ziling Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Shu Song
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Chenguang Wang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China.,Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences & Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, People's Republic of China
| | - Zhengfan Jiang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China.,Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences & Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, People's Republic of China
| | - Youhua Xie
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Zhongtian Qi
- Department of Microbiology, Second Military Medical University, Shanghai, People's Republic of China
| | - Chen Zhao
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Ping Zhao
- Department of Microbiology, Second Military Medical University, Shanghai, People's Republic of China
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Jianqing Xu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
314
|
Kalkeri R, Cai Z, Lin S, Farmer J, Kuzmichev YV, Koide F. SARS-CoV-2 Spike Pseudoviruses: A Useful Tool to Study Virus Entry and Address Emerging Neutralization Escape Phenotypes. Microorganisms 2021; 9:1744. [PMID: 34442823 PMCID: PMC8398529 DOI: 10.3390/microorganisms9081744] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 12/12/2022] Open
Abstract
SARS-CoV-2 genetic variants are emerging around the globe. Unfortunately, several SARS-CoV-2 variants, especially variants of concern (VOCs), are less susceptible to neutralization by the convalescent and post-vaccination sera, raising concerns of increased disease transmissibility and severity. Recent data suggests that SARS-CoV-2 neutralizing antibody levels are a reliable correlate of vaccine-mediated protection. However, currently used BSL3-based virus micro-neutralization (MN) assays are more laborious, time-consuming, and expensive, underscoring the need for BSL2-based, cost-effective neutralization assays against SARS-CoV-2 variants. In light of this unmet need, we have developed a BSL-2 pseudovirus-based neutralization assay (PBNA) in cells expressing the human angiotensin-converting enzyme-2 (hACE2) receptor for SARS-CoV-2. The assay is reproducible (R2 = 0.96), demonstrates a good dynamic range and high sensitivity. Our data suggest that the biological Anti-SARS-CoV-2 research reagents such as NIBSC 20/130 show lower neutralization against B.1.351 SA (South Africa) and B.1.1.7 UK (United Kingdom) VOC, whereas a commercially available monoclonal antibody MM43 retains activity against both these variants. SARS-CoV-2 spike PBNAs for VOCs would be useful tools to measure the neutralization ability of candidate vaccines in both preclinical models and clinical trials and would further help develop effective prophylactic countermeasures against emerging neutralization escape phenotypes.
Collapse
Affiliation(s)
- Raj Kalkeri
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| | - Zhaohui Cai
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| | - Shuling Lin
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| | - John Farmer
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 2000 Ninth Avenue South, Birmingham, AL 35205, USA;
| | - Yury V. Kuzmichev
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| | - Fusataka Koide
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| |
Collapse
|
315
|
Muecksch F, Weisblum Y, Barnes CO, Schmidt F, Schaefer-Babajew D, Wang Z, C Lorenzi JC, Flyak AI, DeLaitsch AT, Huey-Tubman KE, Hou S, Schiffer CA, Gaebler C, Da Silva J, Poston D, Finkin S, Cho A, Cipolla M, Oliveira TY, Millard KG, Ramos V, Gazumyan A, Rutkowska M, Caskey M, Nussenzweig MC, Bjorkman PJ, Hatziioannou T, Bieniasz PD. Affinity maturation of SARS-CoV-2 neutralizing antibodies confers potency, breadth, and resilience to viral escape mutations. Immunity 2021; 54:1853-1868.e7. [PMID: 34331873 PMCID: PMC8323339 DOI: 10.1016/j.immuni.2021.07.008] [Citation(s) in RCA: 208] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/26/2021] [Accepted: 07/12/2021] [Indexed: 12/23/2022]
Abstract
Antibodies elicited by infection accumulate somatic mutations in germinal centers that can increase affinity for cognate antigens. We analyzed 6 independent groups of clonally related severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) Spike receptor-binding domain (RBD)-specific antibodies from 5 individuals shortly after infection and later in convalescence to determine the impact of maturation over months. In addition to increased affinity and neutralization potency, antibody evolution changed the mutational pathways for the acquisition of viral resistance and restricted neutralization escape options. For some antibodies, maturation imposed a requirement for multiple substitutions to enable escape. For certain antibodies, affinity maturation enabled the neutralization of circulating SARS-CoV-2 variants of concern and heterologous sarbecoviruses. Antibody-antigen structures revealed that these properties resulted from substitutions that allowed additional variability at the interface with the RBD. These findings suggest that increasing antibody diversity through prolonged or repeated antigen exposure may improve protection against diversifying SARS-CoV-2 populations, and perhaps against other pandemic threat coronaviruses.
Collapse
MESH Headings
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/chemistry
- Antibodies, Viral/immunology
- Antibody Affinity/immunology
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- COVID-19/immunology
- COVID-19/virology
- Epitopes/chemistry
- Epitopes/immunology
- Host-Pathogen Interactions/immunology
- Humans
- Models, Molecular
- Mutation
- Neutralization Tests
- Protein Binding
- Protein Conformation
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- SARS-CoV-2/pathogenicity
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/immunology
- Structure-Activity Relationship
- Virulence/genetics
Collapse
Affiliation(s)
- Frauke Muecksch
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065, USA
| | - Yiska Weisblum
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065, USA
| | - Christopher O Barnes
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Fabian Schmidt
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065, USA
| | | | - Zijun Wang
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Julio C C Lorenzi
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Andrew I Flyak
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Andrew T DeLaitsch
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Kathryn E Huey-Tubman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Shurong Hou
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Celia A Schiffer
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Christian Gaebler
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Justin Da Silva
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065, USA
| | - Daniel Poston
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065, USA
| | - Shlomo Finkin
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Alice Cho
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Melissa Cipolla
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Katrina G Millard
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Victor Ramos
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Anna Gazumyan
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Magdalena Rutkowska
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065, USA
| | - Marina Caskey
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute.
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| | | | - Paul D Bieniasz
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute.
| |
Collapse
|
316
|
Sampson AT, Heeney J, Cantoni D, Ferrari M, Sans MS, George C, Di Genova C, Mayora Neto M, Einhauser S, Asbach B, Wagner R, Baxendale H, Temperton N, Carnell G. Coronavirus Pseudotypes for All Circulating Human Coronaviruses for Quantification of Cross-Neutralizing Antibody Responses. Viruses 2021; 13:1579. [PMID: 34452443 PMCID: PMC8402765 DOI: 10.3390/v13081579] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/17/2021] [Accepted: 08/01/2021] [Indexed: 12/23/2022] Open
Abstract
The novel coronavirus SARS-CoV-2 is the seventh identified human coronavirus. Understanding the extent of pre-existing immunity induced by seropositivity to endemic seasonal coronaviruses and the impact of cross-reactivity on COVID-19 disease progression remains a key research question in immunity to SARS-CoV-2 and the immunopathology of COVID-2019 disease. This paper describes a panel of lentiviral pseudotypes bearing the spike (S) proteins for each of the seven human coronaviruses (HCoVs), generated under similar conditions optimized for high titre production allowing a high-throughput investigation of antibody neutralization breadth. Optimal production conditions and most readily available permissive target cell lines were determined for spike-mediated entry by each HCoV pseudotype: SARS-CoV-1, SARS-CoV-2 and HCoV-NL63 best transduced HEK293T/17 cells transfected with ACE2 and TMPRSS2, HCoV-229E and MERS-CoV preferentially entered HUH7 cells, and CHO cells were most permissive for the seasonal betacoronavirus HCoV-HKU1. Entry of ACE2 using pseudotypes was enhanced by ACE2 and TMPRSS2 expression in target cells, whilst TMPRSS2 transfection rendered HEK293T/17 cells permissive for HCoV-HKU1 and HCoV-OC43 entry. Additionally, pseudotype viruses were produced bearing additional coronavirus surface proteins, including the SARS-CoV-2 Envelope (E) and Membrane (M) proteins and HCoV-OC43/HCoV-HKU1 Haemagglutinin-Esterase (HE) proteins. This panel of lentiviral pseudotypes provides a safe, rapidly quantifiable and high-throughput tool for serological comparison of pan-coronavirus neutralizing responses; this can be used to elucidate antibody dynamics against individual coronaviruses and the effects of antibody cross-reactivity on clinical outcome following natural infection or vaccination.
Collapse
Affiliation(s)
- Alexander Thomas Sampson
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (J.H.); (M.F.); (M.S.S.); (C.G.); (G.C.)
| | - Jonathan Heeney
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (J.H.); (M.F.); (M.S.S.); (C.G.); (G.C.)
- DIOSynVax Ltd., Cambridge CB3 0ES, UK
| | - Diego Cantoni
- Viral Pseudotype Unit, University of Kent, Chatham ME4 4TB, UK; (D.C.); (C.D.G.); (M.M.N.); (N.T.)
| | - Matteo Ferrari
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (J.H.); (M.F.); (M.S.S.); (C.G.); (G.C.)
- DIOSynVax Ltd., Cambridge CB3 0ES, UK
| | - Maria Suau Sans
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (J.H.); (M.F.); (M.S.S.); (C.G.); (G.C.)
| | - Charlotte George
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (J.H.); (M.F.); (M.S.S.); (C.G.); (G.C.)
| | - Cecilia Di Genova
- Viral Pseudotype Unit, University of Kent, Chatham ME4 4TB, UK; (D.C.); (C.D.G.); (M.M.N.); (N.T.)
| | - Martin Mayora Neto
- Viral Pseudotype Unit, University of Kent, Chatham ME4 4TB, UK; (D.C.); (C.D.G.); (M.M.N.); (N.T.)
| | - Sebastian Einhauser
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany; (S.E.); (B.A.); (R.W.)
| | - Benedikt Asbach
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany; (S.E.); (B.A.); (R.W.)
| | - Ralf Wagner
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany; (S.E.); (B.A.); (R.W.)
- Institute for Clinical Microbiology and Hygiene, University Hospital, 93053 Regensburg, Germany
| | - Helen Baxendale
- Royal Papworth Hospital NHS Foundation Trust, Cambridge CB2 0AY, UK;
| | - Nigel Temperton
- Viral Pseudotype Unit, University of Kent, Chatham ME4 4TB, UK; (D.C.); (C.D.G.); (M.M.N.); (N.T.)
| | - George Carnell
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (J.H.); (M.F.); (M.S.S.); (C.G.); (G.C.)
| |
Collapse
|
317
|
Immunogenicity and protective efficacy of an intranasal live-attenuated vaccine against SARS-CoV-2. iScience 2021; 24:102941. [PMID: 34368648 PMCID: PMC8332743 DOI: 10.1016/j.isci.2021.102941] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 06/27/2021] [Accepted: 07/30/2021] [Indexed: 01/08/2023] Open
Abstract
Global deployment of an effective and safe vaccine is necessary to curtail the coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here, we evaluated a Newcastle disease virus (NDV)-based vectored-vaccine in mice and hamsters for its immunogenicity, safety, and protective efficacy against SARS-CoV-2. Intranasal administration of recombinant (r)NDV-S vaccine expressing spike (S) protein of SARS-CoV-2 to mice induced high levels of SARS-CoV-2-specific neutralizing immunoglobulin A (IgA) and IgG2a antibodies and T-cell-mediated immunity. Hamsters immunized with two doses of vaccine showed complete protection from lung infection, inflammation, and pathological lesions following SARS-CoV-2 challenge. Importantly, administration of two doses of intranasal rNDV-S vaccine significantly reduced the SARS-CoV-2 shedding in nasal turbinate and lungs in hamsters. Collectively, intranasal vaccination has the potential to control infection at the site of inoculation, which should prevent both clinical disease and virus transmission to halt the spread of the COVID-19 pandemic. Vaccine induces high levels of neutralizing Abs and T-cell-mediated immunity Vaccine ameliorates lung inflammation and pathology in hamster induced by SARS-CoV-2 The SARS-CoV-2 remains undetectable in lungs and nasal turbinates of vaccinated hamster Two doses of intranasal vaccine show complete protection against SARS-CoV-2 challenge
Collapse
|
318
|
Li W, Chen Y, Prévost J, Ullah I, Lu M, Gong SY, Tauzin A, Gasser R, Vézina D, Anand SP, Goyette G, Chaterjee D, Ding S, Tolbert WD, Grunst MW, Bo Y, Zhang S, Richard J, Zhou F, Huang RK, Esser L, Zeher A, Côté M, Kumar P, Sodroski J, Xia D, Uchil PD, Pazgier M, Finzi A, Mothes W. Structural Basis and Mode of Action for Two Broadly Neutralizing Antibodies Against SARS-CoV-2 Emerging Variants of Concern. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.08.02.454546. [PMID: 34373853 PMCID: PMC8351775 DOI: 10.1101/2021.08.02.454546] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Emerging variants of concern for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can transmit more efficiently and partially evade protective immune responses, thus necessitating continued refinement of antibody therapies and immunogen design. Here we elucidate the structural basis and mode of action for two potent SARS-CoV-2 Spike (S) neutralizing monoclonal antibodies CV3-1 and CV3-25 that remained effective against emerging variants of concern in vitro and in vivo. CV3-1 bound to the (485-GFN-487) loop within the receptor-binding domain (RBD) in the "RBD-up" position and triggered potent shedding of the S1 subunit. In contrast, CV3-25 inhibited membrane fusion by binding to an epitope in the stem helix region of the S2 subunit that is highly conserved among β-coronaviruses. Thus, vaccine immunogen designs that incorporate the conserved regions in RBD and stem helix region are candidates to elicit pan-coronavirus protective immune responses.
Collapse
Affiliation(s)
- Wenwei Li
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Yaozong Chen
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814-4712, USA
| | - Jérémie Prévost
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Irfan Ullah
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520
| | - Maolin Lu
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Shang Yu Gong
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Alexandra Tauzin
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Romain Gasser
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Dani Vézina
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Sai Priya Anand
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | | | | | - Shilei Ding
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - William D. Tolbert
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814-4712, USA
| | - Michael W. Grunst
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Yuxia Bo
- Department of Biochemistry, Microbiology and Immunology, and Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Shijian Zhang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Jonathan Richard
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Fei Zhou
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rick K. Huang
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lothar Esser
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Allison Zeher
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, and Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Priti Kumar
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520
| | - Joseph Sodroski
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Di Xia
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pradeep D. Uchil
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Marzena Pazgier
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814-4712, USA
| | - Andrés Finzi
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Walther Mothes
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
319
|
Alenquer M, Ferreira F, Lousa D, Valério M, Medina-Lopes M, Bergman ML, Gonçalves J, Demengeot J, Leite RB, Lilue J, Ning Z, Penha-Gonçalves C, Soares H, Soares CM, Amorim MJ. Signatures in SARS-CoV-2 spike protein conferring escape to neutralizing antibodies. PLoS Pathog 2021; 17:e1009772. [PMID: 34352039 PMCID: PMC8341613 DOI: 10.1371/journal.ppat.1009772] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
Understanding SARS-CoV-2 evolution and host immunity is critical to control COVID-19 pandemics. At the core is an arms-race between SARS-CoV-2 antibody and angiotensin-converting enzyme 2 (ACE2) recognition, a function of the viral protein spike. Mutations in spike impacting antibody and/or ACE2 binding are appearing worldwide, imposing the need to monitor SARS-CoV2 evolution and dynamics in the population. Determining signatures in SARS-CoV-2 that render the virus resistant to neutralizing antibodies is critical. We engineered 25 spike-pseudotyped lentiviruses containing individual and combined mutations in the spike protein, including all defining mutations in the variants of concern, to identify the effect of single and synergic amino acid substitutions in promoting immune escape. We confirmed that E484K evades antibody neutralization elicited by infection or vaccination, a capacity augmented when complemented by K417N and N501Y mutations. In silico analysis provided an explanation for E484K immune evasion. E484 frequently engages in interactions with antibodies but not with ACE2. Importantly, we identified a novel amino acid of concern, S494, which shares a similar pattern. Using the already circulating mutation S494P, we found that it reduces antibody neutralization of convalescent and post-immunization sera, particularly when combined with E484K and with mutations able to increase binding to ACE2, such as N501Y. Our analysis of synergic mutations provides a signature for hotspots for immune evasion and for targets of therapies, vaccines and diagnostics.
Collapse
Affiliation(s)
- Marta Alenquer
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência; Oeiras, Portugal
| | - Filipe Ferreira
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência; Oeiras, Portugal
| | - Diana Lousa
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa; Oeiras, Portugal
| | - Mariana Valério
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa; Oeiras, Portugal
| | - Mónica Medina-Lopes
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência; Oeiras, Portugal
| | | | - Juliana Gonçalves
- CEDOC NOVA, Centro de Estudos de Doenças Crónicas, Nova Medical School, Universidade Nova de Lisboa; Lisboa, Portugal
| | - Jocelyne Demengeot
- Lymphocyte Physiology Lab, Instituto Gulbenkian de Ciência; Oeiras, Portugal
| | - Ricardo B. Leite
- Genomics Unit, Instituto Gulbenkian de Ciência; Oeiras, Portugal
| | - Jingtao Lilue
- Bioinformatics Unit, Instituto Gulbenkian de Ciência; Oeiras, Portugal
| | - Zemin Ning
- The Wellcome Trust Sanger Institute; Hinxton, United Kingdom
| | | | - Helena Soares
- CEDOC NOVA, Centro de Estudos de Doenças Crónicas, Nova Medical School, Universidade Nova de Lisboa; Lisboa, Portugal
| | - Cláudio M. Soares
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa; Oeiras, Portugal
| | - Maria João Amorim
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência; Oeiras, Portugal
| |
Collapse
|
320
|
Collier DA, Ferreira IATM, Kotagiri P, Datir RP, Lim EY, Touizer E, Meng B, Abdullahi A, Elmer A, Kingston N, Graves B, Le Gresley E, Caputo D, Bergamaschi L, Smith KGC, Bradley JR, Ceron-Gutierrez L, Cortes-Acevedo P, Barcenas-Morales G, Linterman MA, McCoy LE, Davis C, Thomson E, Lyons PA, McKinney E, Doffinger R, Wills M, Gupta RK. Age-related immune response heterogeneity to SARS-CoV-2 vaccine BNT162b2. Nature 2021; 596:417-422. [PMID: 34192737 PMCID: PMC8373615 DOI: 10.1038/s41586-021-03739-1] [Citation(s) in RCA: 505] [Impact Index Per Article: 126.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/18/2021] [Indexed: 12/18/2022]
Abstract
Although two-dose mRNA vaccination provides excellent protection against SARS-CoV-2, there is little information about vaccine efficacy against variants of concern (VOC) in individuals above eighty years of age1. Here we analysed immune responses following vaccination with the BNT162b2 mRNA vaccine2 in elderly participants and younger healthcare workers. Serum neutralization and levels of binding IgG or IgA after the first vaccine dose were lower in older individuals, with a marked drop in participants over eighty years old. Sera from participants above eighty showed lower neutralization potency against the B.1.1.7 (Alpha), B.1.351 (Beta) and P.1. (Gamma) VOC than against the wild-type virus and were more likely to lack any neutralization against VOC following the first dose. However, following the second dose, neutralization against VOC was detectable regardless of age. The frequency of SARS-CoV-2 spike-specific memory B cells was higher in elderly responders (whose serum showed neutralization activity) than in non-responders after the first dose. Elderly participants showed a clear reduction in somatic hypermutation of class-switched cells. The production of interferon-γ and interleukin-2 by SARS-CoV-2 spike-specific T cells was lower in older participants, and both cytokines were secreted primarily by CD4 T cells. We conclude that the elderly are a high-risk population and that specific measures to boost vaccine responses in this population are warranted, particularly where variants of concern are circulating.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Aging/blood
- Aging/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Autoantibodies/immunology
- B-Lymphocytes/cytology
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- BNT162 Vaccine
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/immunology
- Female
- Health Personnel
- Humans
- Immunity/genetics
- Immunization, Secondary
- Immunoglobulin A/immunology
- Immunoglobulin Class Switching
- Immunoglobulin G/genetics
- Immunoglobulin G/immunology
- Immunologic Memory/immunology
- Inflammation/blood
- Inflammation/immunology
- Interferon-gamma/immunology
- Interleukin-2/immunology
- Male
- Middle Aged
- SARS-CoV-2/immunology
- Somatic Hypermutation, Immunoglobulin
- Spike Glycoprotein, Coronavirus/immunology
- T-Lymphocytes/immunology
- Vaccination
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
- mRNA Vaccines
Collapse
Affiliation(s)
- Dami A Collier
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- Division of Infection and Immunity, University College London, London, UK
| | - Isabella A T M Ferreira
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Prasanti Kotagiri
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Rawlings P Datir
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- Division of Infection and Immunity, University College London, London, UK
| | - Eleanor Y Lim
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Emma Touizer
- Division of Infection and Immunity, University College London, London, UK
| | - Bo Meng
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Adam Abdullahi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Anne Elmer
- NIHR Bioresource, Cambridge, UK
- Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Nathalie Kingston
- NIHR Bioresource, Cambridge, UK
- Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Emma Le Gresley
- NIHR Bioresource, Cambridge, UK
- Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Daniela Caputo
- NIHR Bioresource, Cambridge, UK
- Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Laura Bergamaschi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - John R Bradley
- Department of Medicine, University of Cambridge, Cambridge, UK
- NIHR Bioresource, Cambridge, UK
| | | | | | | | | | - Laura E McCoy
- Division of Infection and Immunity, University College London, London, UK
| | - Chris Davis
- MRC Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Emma Thomson
- MRC Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Eoin McKinney
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Rainer Doffinger
- Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
| | - Mark Wills
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
321
|
Scaglione A, Opp S, Hurtado A, Lin Z, Pampeno C, Noval MG, Thannickal SA, Stapleford KA, Meruelo D. Combination of a Sindbis-SARS-CoV-2 Spike Vaccine and αOX40 Antibody Elicits Protective Immunity Against SARS-CoV-2 Induced Disease and Potentiates Long-Term SARS-CoV-2-Specific Humoral and T-Cell Immunity. Front Immunol 2021; 12:719077. [PMID: 34394127 PMCID: PMC8359677 DOI: 10.3389/fimmu.2021.719077] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/13/2021] [Indexed: 12/17/2022] Open
Abstract
The COVID-19 pandemic caused by the coronavirus SARS-CoV-2 is a major global public threat. Currently, a worldwide effort has been mounted to generate billions of effective SARS-CoV-2 vaccine doses to immunize the world's population at record speeds. However, there is still a demand for alternative effective vaccines that rapidly confer long-term protection and rely upon cost-effective, easily scaled-up manufacturing. Here, we present a Sindbis alphavirus vector (SV), transiently expressing the SARS-CoV-2 spike protein (SV.Spike), combined with the OX40 immunostimulatory antibody (αOX40) as a novel, highly effective vaccine approach. We show that SV.Spike plus αOX40 elicits long-lasting neutralizing antibodies and a vigorous T-cell response in mice. Protein binding, immunohistochemical, and cellular infection assays all show that vaccinated mice sera inhibits spike functions. Immunophenotyping, RNA Seq transcriptome profiles, and metabolic analysis indicate a reprogramming of T cells in vaccinated mice. Activated T cells were found to mobilize to lung tissue. Most importantly, SV.Spike plus αOX40 provided robust immune protection against infection with authentic coronavirus in transgenic mice expressing the human ACE2 receptor (hACE2-Tg). Finally, our immunization strategy induced strong effector memory response, potentiating protective immunity against re-exposure to SARS-CoV-2 spike protein. Our results show the potential of a new Sindbis virus-based vaccine platform to counteract waning immune response, which can be used as a new candidate to combat SARS-CoV-2. Given the T-cell responses elicited, our vaccine is likely to be effective against variants that are proving challenging, as well as serve as a platform to develop a broader spectrum pancoronavirus vaccine. Similarly, the vaccine approach is likely to be applicable to other pathogens.
Collapse
Affiliation(s)
- Antonella Scaglione
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Silvana Opp
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Alicia Hurtado
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Ziyan Lin
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Christine Pampeno
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Maria G. Noval
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, United States
| | - Sara A. Thannickal
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, United States
| | - Kenneth A. Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, United States
| | - Daniel Meruelo
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
322
|
Rapid and Flexible Platform To Assess Anti-SARS-CoV-2 Antibody Neutralization and Spike Protein-Specific Antivirals. mSphere 2021; 6:e0057121. [PMID: 34319126 PMCID: PMC8386372 DOI: 10.1128/msphere.00571-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) is ongoing and has shown the community that flexible methods for rapidly identifying and screening candidate antivirals are needed. Assessing virus-neutralizing activity of human serum to monitor population immunity and response to infection and vaccination is key to pandemic control. We developed a virus neutralization platform strategy that relies only on bioinformatic and genetic information of the virus of interest. The platform uses viral envelope glycoprotein cDNAs to set up an assay that mimics multicycle infection but is safe and, therefore, amenable to biosafety level 2 (BSL2) conditions for viruses that require BSL3 facilities (e.g., SARS-CoV-1 and SARS-CoV-2). As a complement to this platform, we present a new cell-based immunofluorescent (CBI) assay that uses SARS-CoV-2 spike protein (S)-expressing cells to accurately measure the neutralization potential of human sera and is readily adaptable to variants of concern. These methods should be useful additions to the tools for assessing antiviral immunity, whether acquired via natural infection or vaccines. IMPORTANCE Assays for rapid biosafety level 2 (BSL2) evaluation of neutralizing properties of antibodies acquired via natural infection or through vaccination is urgently needed. Here, we propose a combinatorial approach in which sera are screened for SARS-CoV-2 spike protein (S) binding using a cell-based immunofluorescent (CBI) assay, and positive samples are further evaluated in a pseudotyped viral multicycle infection-mimicking protocol under BSL2 conditions.
Collapse
|
323
|
Alsaidi S, Cornejal N, Mahoney O, Melo C, Verma N, Bonnaire T, Chang T, O’Keefe BR, Sailer J, Zydowsky TM, Teleshova N, Romero JAF. Griffithsin and Carrageenan Combination Results in Antiviral Synergy against SARS-CoV-1 and 2 in a Pseudoviral Model. Mar Drugs 2021; 19:md19080418. [PMID: 34436255 PMCID: PMC8400000 DOI: 10.3390/md19080418] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/15/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023] Open
Abstract
Over 182 million confirmed cases of COVID-19 and more than 4 million deaths have been reported to date around the world. It is essential to identify broad-spectrum antiviral agents that may prevent or treat infections by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) but also by other coronaviruses that may jump the species barrier in the future. We evaluated the antiviral selectivity of griffithsin and sulfated and non-sulfated polysaccharides against SARS-CoV-1 and SARS-CoV-2 using a cytotoxicity assay and a cell-based pseudoviral model. The half-maximal cytotoxic concentration (CC50) and half-maximal effective concentration (EC50) were determined for each compound, using a dose-response-inhibition analysis on GraphPad Prism v9.0.2 software (San Diego, CA, USA). The therapeutic index (TI = CC50/EC50) was calculated for each compound. The potential synergistic, additive, or antagonistic effect of different compound combinations was determined by CalcuSyn v1 software (Biosoft, Cambridge, UK), which estimated the combination index (CI) values. Iota and lambda carrageenan showed the most potent antiviral activity (EC50 between 3.2 and 7.5 µg/mL). Carrageenan and griffithsin combinations exhibited synergistic activity (EC50 between 0.2 and 3.8 µg/mL; combination index <1), including against recent SARS-CoV-2 mutations. The griffithsin and carrageenan combination is a promising candidate to prevent or treat infections by SARS-CoV-1 and SARS-CoV-2.
Collapse
Affiliation(s)
- Sahar Alsaidi
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
- Department of Science, Borough of Manhattan Community College, The City University of New York, New York, NY 10007, USA
- Department of Anthropology, Lehman College, The City University of New York, New York, NY 10468, USA
| | - Nadjet Cornejal
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
- Department of Science, Borough of Manhattan Community College, The City University of New York, New York, NY 10007, USA
- Center for Achievement in Science Education, Department of Biology and Chemistry, School of Natural and Behavioral Sciences, Brooklyn College, The City University of New York, New York, NY 11210, USA
| | - Oneil Mahoney
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
- Department of Science, Borough of Manhattan Community College, The City University of New York, New York, NY 10007, USA
| | - Claudia Melo
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
- Department of Science, Borough of Manhattan Community College, The City University of New York, New York, NY 10007, USA
- Center for Achievement in Science Education, Department of Biology and Chemistry, School of Natural and Behavioral Sciences, Brooklyn College, The City University of New York, New York, NY 11210, USA
| | - Neeharika Verma
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
| | - Thierry Bonnaire
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
| | - Theresa Chang
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07102, USA;
| | - Barry R. O’Keefe
- Natural Products Branch, Molecular Targets Program, Developmental Therapeutics Program, Center for Cancer Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD 21702, USA;
| | - James Sailer
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
| | - Thomas M. Zydowsky
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
| | - Natalia Teleshova
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
| | - José A. Fernández Romero
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
- Department of Science, Borough of Manhattan Community College, The City University of New York, New York, NY 10007, USA
- Correspondence:
| |
Collapse
|
324
|
Systemic IL-15, IFN-γ, and IP-10/CXCL10 signature associated with effective immune response to SARS-CoV-2 in BNT162b2 mRNA vaccine recipients. Cell Rep 2021; 36:109504. [PMID: 34352226 PMCID: PMC8299183 DOI: 10.1016/j.celrep.2021.109504] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/16/2021] [Accepted: 07/19/2021] [Indexed: 12/29/2022] Open
Abstract
Early responses to vaccination are important for shaping both humoral and cellular protective immunity. Dissecting innate vaccine signatures may predict immunogenicity to help optimize the efficacy of mRNA and other vaccine strategies. Here, we characterize the cytokine and chemokine responses to the 1st and 2nd dose of the BNT162b2 mRNA (Pfizer/BioNtech) vaccine in antigen-naive and in previously coronavirus disease 2019 (COVID-19)-infected individuals (NCT04743388). Transient increases in interleukin-15 (IL-15) and interferon gamma (IFN-γ) levels early after boost correlate with Spike antibody levels, supporting their use as biomarkers of effective humoral immunity development in response to vaccination. We identify a systemic signature including increases in IL-15, IFN-γ, and IP-10/CXCL10 after the 1st vaccination, which were enriched by tumor necrosis factor alpha (TNF-α) and IL-6 after the 2nd vaccination. In previously COVID-19-infected individuals, a single vaccination results in both strong cytokine induction and antibody titers similar to the ones observed upon booster vaccination in antigen-naive individuals, a result with potential implication for future public health recommendations.
Collapse
|
325
|
Performance of Three SARS-CoV-2 Immunoassays, Three Rapid Lateral Flow Tests, and a Novel Bead-Based Affinity Surrogate Test for the Detection of SARS-CoV-2 Antibodies in Human Serum. J Clin Microbiol 2021; 59:e0031921. [PMID: 33962959 DOI: 10.1128/jcm.00319-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
For the control of immunity in COVID-19 survivors and vaccinated subjects, there is an urgent need for reliable and rapid serological assays. Based on samples from 63 COVID-19 survivors up to 7 months after symptom onset, and on 50 serum samples taken before the beginning of the pandemic, we compared the performances of three commercial immunoassays for the detection of SARS-CoV-2 IgA and IgG antibodies (Euroimmun SARS-COV-2 IgA/IgG, Mikrogen recomWell SARS-CoV-2 IgA/IgG, and Serion ELISA agile SARS-CoV-2 IgA/IgG) and three rapid lateral flow (immunochromatographic) tests (Abbott PanBio COVID-19 IgG/IgM, Nadal COVID-19 IgG/IgM, and Cleartest Corona 2019-nCOV IgG/IgM) with a 50% plaque-reduction neutralization test (PRNT50) representing the gold standard. Fifty-seven out of 63 PCR-confirmed COVID-19 patients (90%) showed neutralizing antibodies. The sensitivity of the seven assays ranged from 7.0% to 98.3%, and the specificity ranged from 86.0% to 100.0%. Only one commercial immunoassay showed a sensitivity and specificity of greater than 98%.
Collapse
|
326
|
Boppana S, Qin K, Files JK, Russell RM, Stoltz R, Bibollet-Ruche F, Bansal A, Erdmann N, Hahn BH, Goepfert PA. SARS-CoV-2-specific circulating T follicular helper cells correlate with neutralizing antibodies and increase during early convalescence. PLoS Pathog 2021; 17:e1009761. [PMID: 34270631 PMCID: PMC8318272 DOI: 10.1371/journal.ppat.1009761] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/28/2021] [Accepted: 06/27/2021] [Indexed: 01/08/2023] Open
Abstract
T-cell immunity is likely to play a role in protection against SARS-CoV-2 by helping generate neutralizing antibodies. We longitudinally studied CD4 T-cell responses to the M, N, and S structural proteins of SARS-CoV-2 in 26 convalescent individuals. Within the first two months following symptom onset, a majority of individuals (81%) mounted at least one CD4 T-cell response, and 48% of individuals mounted detectable SARS-CoV-2-specific circulating T follicular helper cells (cTfh, defined as CXCR5+PD1+ CD4 T cells). SARS-CoV-2-specific cTfh responses across all three protein specificities correlated with antibody neutralization with the strongest correlation observed for S protein-specific responses. When examined over time, cTfh responses, particularly to the M protein, increased in convalescence, and robust cTfh responses with magnitudes greater than 5% were detected at the second convalescent visit, a median of 38 days post-symptom onset. CD4 T-cell responses declined but persisted at low magnitudes three months and six months after symptom onset. These data deepen our understanding of antigen-specific cTfh responses in SARS-CoV-2 infection, suggesting that in addition to S protein, M and N protein-specific cTfh may also assist in the development of neutralizing antibodies and that cTfh response formation may be delayed in SARS-CoV-2 infection. Since December 2019, the Coronavirus Disease 2019 (COVID-19) pandemic has caused significant morbidity and mortality worldwide. Recently approved vaccines against SARS-CoV-2 are understood to protect against infection by inducing neutralizing antibodies. However, the underlying immune responses necessary for protection remain unclear. It is well established that T follicular helper cells (Tfh), a subset of CD4 T cells, are essential to the development of neutralizing antibodies and that some of these cells, called circulating T follicular helper cells (cTfh), can be studied in the blood. Not much is known about Tfh responses mounted in SARS-CoV-2 infection. Here, we studied cTfh responses to three major structural proteins in individuals recovered from COVID-19. We find that SARS-CoV-2-specific cTfh frequencies correlate with neutralizing antibody responses. We also find that cTfh responses to SARS-CoV-2 increase well into convalescence before contracting. Our results suggest that cTfh responses against proteins other than the spike protein may contribute to the development of neutralizing antibodies and that the formation of cTfh responses in SARS-CoV-2 infection may be delayed.
Collapse
Affiliation(s)
- Sushma Boppana
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Kai Qin
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jacob K Files
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ronnie M Russell
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America.,Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Regina Stoltz
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Frederic Bibollet-Ruche
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America.,Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Anju Bansal
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Nathan Erdmann
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Beatrice H Hahn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America.,Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Paul A Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
327
|
Muecksch F, Wise H, Templeton K, Batchelor B, Squires M, McCance K, Jarvis L, Malloy K, Furrie E, Richardson C, MacGuire J, Godber I, Burns A, Mavin S, Zhang F, Schmidt F, Bieniasz P, Jenks S, Hatziioannou T. Longitudinal variation in SARS-CoV-2 antibody levels and emergence of viral variants: implications for the ability of serological assays to predict immunity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.07.02.21259939. [PMID: 34268524 PMCID: PMC8282113 DOI: 10.1101/2021.07.02.21259939] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Serological assays are being deployed to monitor antibody responses in SARS-CoV-2 convalescents and vaccine recipients. There is a need to determine whether such assays can predict immunity, as antibody levels wane and viral variants emerge. METHODS We measured antibodies in a cohort of SARS-CoV-2 infected patients using several high-throughput serological tests and functional neutralization assays. The effects of time and spike protein sequence variation on the performance and predictive value of the various assays was assessed. FINDINGS Neutralizing antibody titers decreased over the first few months post-infection but stabilized thereafter, at about 30% of the level observed shortly after infection. Serological assays commonly used to measure antibodies against SARS-CoV-2 displayed a range of sensitivities that declined to varying extents over time. Quantitative measurements generated by serological assays based on the spike protein were better at predicting neutralizing antibody titers than assays based on nucleocapsid, but performance was variable and manufacturer positivity thresholds were not able to predict the presence or absence of detectable neutralizing activity. Even though there was some deterioration in correlation between serological measurements and functional neutralization activity, some assays maintained an ability to predict neutralizing titers, even against variants of concern. INTERPRETATION The ability of high throughput serological assays to predict neutralizing antibody titers is likely crucial for evaluation of immunity at the population scale. These data will facilitate the selection of the most suitable assays as surrogates of functional neutralizing activity and suggest that such measurements may have utility in clinical practice.
Collapse
Affiliation(s)
- Frauke Muecksch
- Laboratory of Retrovirology, The Rockefeller University, 1230 York Avenue, New York NY 10065
| | - Helen Wise
- Royal Infirmary of Edinburgh, NHS Lothian, 51 Little France Crescent, Edinburgh EH16 4SA
| | - Kate Templeton
- Royal Infirmary of Edinburgh, NHS Lothian, 51 Little France Crescent, Edinburgh EH16 4SA
| | - Becky Batchelor
- Western General Hospital, Crewe Road South, Edinburgh, EH4 2XU
| | - Maria Squires
- Royal Infirmary of Edinburgh, NHS Lothian, 51 Little France Crescent, Edinburgh EH16 4SA
| | - Kirsty McCance
- Royal Infirmary of Edinburgh, NHS Lothian, 51 Little France Crescent, Edinburgh EH16 4SA
| | - Lisa Jarvis
- SNBTS Microbiology Reference Laboratory, The Jack Copland Centre, 52 Research Avenue North, Heriot Watt Research Park, Edinburgh, EH14 4AP
| | - Kristen Malloy
- SNBTS Microbiology Reference Laboratory, The Jack Copland Centre, 52 Research Avenue North, Heriot Watt Research Park, Edinburgh, EH14 4AP
| | - Elizabeth Furrie
- Ninewells Hospital and Medical School, NHS Tayside, Dundee, DD1 9SY
| | | | | | - Ian Godber
- Queen Elizabeth University Hospital, Govan Road, Glasgow, G51 4TF
| | - Alana Burns
- Queen Elizabeth University Hospital, Govan Road, Glasgow, G51 4TF
| | - Sally Mavin
- Scottish Microbiology Reference Laboratory, NHS Highland, Inverness, IV2 3UJ
| | - Fengwen Zhang
- Laboratory of Retrovirology, The Rockefeller University, 1230 York Avenue, New York NY 10065
| | - Fabian Schmidt
- Laboratory of Retrovirology, The Rockefeller University, 1230 York Avenue, New York NY 10065
| | - Paul Bieniasz
- Laboratory of Retrovirology, The Rockefeller University, 1230 York Avenue, New York NY 10065
- Howard Hughes Medical Institute, The Rockefeller University, 1230 York Avenue, New York NY 10065
| | - Sara Jenks
- Royal Infirmary of Edinburgh, NHS Lothian, 51 Little France Crescent, Edinburgh EH16 4SA
| | - Theodora Hatziioannou
- Laboratory of Retrovirology, The Rockefeller University, 1230 York Avenue, New York NY 10065
| |
Collapse
|
328
|
Xu J, Xu K, Jung S, Conte A, Lieberman J, Muecksch F, Lorenzi JCC, Park S, Schmidt F, Wang Z, Huang Y, Luo Y, Nair MS, Wang P, Schulz JE, Tessarollo L, Bylund T, Chuang GY, Olia AS, Stephens T, Teng IT, Tsybovsky Y, Zhou T, Munster V, Ho DD, Hatziioannou T, Bieniasz PD, Nussenzweig MC, Kwong PD, Casellas R. Nanobodies from camelid mice and llamas neutralize SARS-CoV-2 variants. Nature 2021; 595:278-282. [PMID: 34098567 PMCID: PMC8260353 DOI: 10.1038/s41586-021-03676-z] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/27/2021] [Indexed: 11/08/2022]
Abstract
Since the start of the COVID-19 pandemic, SARS-CoV-2 has caused millions of deaths worldwide. Although a number of vaccines have been deployed, the continual evolution of the receptor-binding domain (RBD) of the virus has challenged their efficacy. In particular, the emerging variants B.1.1.7, B.1.351 and P.1 (first detected in the UK, South Africa and Brazil, respectively) have compromised the efficacy of sera from patients who have recovered from COVID-19 and immunotherapies that have received emergency use authorization1-3. One potential alternative to avert viral escape is the use of camelid VHHs (variable heavy chain domains of heavy chain antibody (also known as nanobodies)), which can recognize epitopes that are often inaccessible to conventional antibodies4. Here, we isolate anti-RBD nanobodies from llamas and from mice that we engineered to produce VHHs cloned from alpacas, dromedaries and Bactrian camels. We identified two groups of highly neutralizing nanobodies. Group 1 circumvents antigenic drift by recognizing an RBD region that is highly conserved in coronaviruses but rarely targeted by human antibodies. Group 2 is almost exclusively focused to the RBD-ACE2 interface and does not neutralize SARS-CoV-2 variants that carry E484K or N501Y substitutions. However, nanobodies in group 2 retain full neutralization activity against these variants when expressed as homotrimers, and-to our knowledge-rival the most potent antibodies against SARS-CoV-2 that have been produced to date. These findings suggest that multivalent nanobodies overcome SARS-CoV-2 mutations through two separate mechanisms: enhanced avidity for the ACE2-binding domain and recognition of conserved epitopes that are largely inaccessible to human antibodies. Therefore, although new SARS-CoV-2 mutants will continue to emerge, nanobodies represent promising tools to prevent COVID-19 mortality when vaccines are compromised.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/isolation & purification
- CRISPR-Cas Systems
- Camelids, New World/genetics
- Camelids, New World/immunology
- Female
- Gene Editing
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Models, Molecular
- Mutation
- Neutralization Tests
- SARS-CoV-2/chemistry
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- Single-Domain Antibodies/chemistry
- Single-Domain Antibodies/genetics
- Single-Domain Antibodies/immunology
- Single-Domain Antibodies/isolation & purification
- Somatic Hypermutation, Immunoglobulin/genetics
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/immunology
Collapse
Affiliation(s)
- Jianliang Xu
- Lymphocyte Nuclear Biology, NIAMS, NIH, Bethesda, MD, USA.
| | - Kai Xu
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Andrea Conte
- Lymphocyte Nuclear Biology, NIAMS, NIH, Bethesda, MD, USA
| | | | - Frauke Muecksch
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | | | - Solji Park
- Lymphocyte Nuclear Biology, NIAMS, NIH, Bethesda, MD, USA
| | - Fabian Schmidt
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | - Zijun Wang
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Yaoxing Huang
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Yang Luo
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Manoj S Nair
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Pengfei Wang
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Jonathan E Schulz
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Lino Tessarollo
- Mouse Cancer Genetics Program, CCR, NCI, NIH, Frederick, MD, USA
| | | | - Gwo-Yu Chuang
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Adam S Olia
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Tyler Stephens
- Electron Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA
| | - I-Ting Teng
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Yaroslav Tsybovsky
- Electron Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA
| | - Tongqing Zhou
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Vincent Munster
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | | | - Paul D Bieniasz
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA.
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA.
| | - Peter D Kwong
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA.
| | - Rafael Casellas
- Lymphocyte Nuclear Biology, NIAMS, NIH, Bethesda, MD, USA.
- The NIH Regulome Project, NIH, Bethesda, MD, USA.
- Center for Cancer Research, NCI, NIH, Bethesda, MD, USA.
| |
Collapse
|
329
|
Muir L, Jaffer A, Rees-Spear C, Gopalan V, Chang FY, Fernando R, Vaitkute G, Roustan C, Rosa A, Earl C, Rajakaruna GK, Cherepanov P, Salama A, McCoy LE, Motallebzadeh R. Neutralizing Antibody Responses After SARS-CoV-2 Infection in End-Stage Kidney Disease and Protection Against Reinfection. Kidney Int Rep 2021; 6:1799-1809. [PMID: 33942026 PMCID: PMC8081267 DOI: 10.1016/j.ekir.2021.03.902] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Patients with end-stage kidney disease (ESKD) represent a vulnerable group with multiple risk factors that are associated with poor outcomes after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Despite established susceptibility to infectious complications and the importance of humoral immunity in protection against SARS-CoV-2, few studies have investigated the humoral immune response to SARS-CoV-2 within this population. Here, we evaluate the seroprevalence of SARS-CoV-2 in patients awaiting renal transplantation and determine whether seroconverted patients with ESKD have durable and functional neutralizing activity against SARS-CoV-2. METHODS Serum samples were obtained from 164 patients with ESKD by August 2020. Humoral immune responses were evaluated by SARS-CoV-2 spike S1 subunit and nucleoprotein semiquantitative enzyme-linked immunosorbent assay (ELISA) and SARS-CoV-2 spike pseudotype neutralization assay. RESULTS All patients with ESKD with reverse-transcriptase polymerase chain reaction (RT-PCR)-confirmed infection (n = 17) except for 1 individual seroconverted against SARS-CoV-2. Overall seroprevalence (anti-S1 and/or anti-N IgG) was 36% and was higher in patients on hemodialysis (44.2%). A total of 35.6% of individuals who seroconverted were asymptomatic. Seroconversion in the absence of a neutralizing antibody (nAb) titer was observed in 12 patients, all of whom were asymptomatic. Repeat measurements at a median of 93 days from baseline sampling revealed that most individuals retained detectable responses although a significant drop in S1, N and nAb titers was observed. CONCLUSION Patients with ESKD, including those who develop asymptomatic disease, routinely seroconvert and produce detectable nAb titers against SARS-CoV-2. Although IgG levels wane over time, the neutralizing antibodies remain detectable in most patients, suggesting some level of protection is likely maintained, particularly in those who originally develop stronger responses.
Collapse
Affiliation(s)
- Luke Muir
- UCL Institute of Immunity & Transplantation, University College London, London, UK
- UCL Division of Infection & Immunity, University College London, London, UK
| | - Aneesa Jaffer
- Department of Nephrology & Transplantation, Royal Free London NHS Trust, London, UK
| | - Chloe Rees-Spear
- UCL Institute of Immunity & Transplantation, University College London, London, UK
- UCL Division of Infection & Immunity, University College London, London, UK
| | - Vignesh Gopalan
- Department of Nephrology & Transplantation, Royal Free London NHS Trust, London, UK
| | - Fernando Y. Chang
- Research Department of Surgical Biotechnology, UCL Division of Surgery and Interventional Science, University College London, London, UK
| | - Raymond Fernando
- Department of Nephrology & Transplantation, Royal Free London NHS Trust, London, UK
| | - Gintare Vaitkute
- Research Department of Surgical Biotechnology, UCL Division of Surgery and Interventional Science, University College London, London, UK
| | | | | | | | - Gayathri K. Rajakaruna
- Centre for Transplantation, Department of Renal Medicine, University College London, London, UK
| | | | - Alan Salama
- Department of Nephrology & Transplantation, Royal Free London NHS Trust, London, UK
- Centre for Transplantation, Department of Renal Medicine, University College London, London, UK
| | - Laura E. McCoy
- UCL Institute of Immunity & Transplantation, University College London, London, UK
- UCL Division of Infection & Immunity, University College London, London, UK
| | - Reza Motallebzadeh
- UCL Institute of Immunity & Transplantation, University College London, London, UK
- Department of Nephrology & Transplantation, Royal Free London NHS Trust, London, UK
- Research Department of Surgical Biotechnology, UCL Division of Surgery and Interventional Science, University College London, London, UK
- Centre for Transplantation, Department of Renal Medicine, University College London, London, UK
| |
Collapse
|
330
|
Li Y, Schneider AM, Mehta A, Sade-Feldman M, Kays KR, Gentili M, Charland NC, Gonye AL, Gushterova I, Khanna HK, LaSalle TJ, Lavin-Parsons KM, Lilley BM, Lodenstein CL, Manakongtreecheep K, Margolin JD, McKaig BN, Parry BA, Rojas-Lopez M, Russo BC, Sharma N, Tantivit J, Thomas MF, Regan J, Flynn JP, Villani AC, Hacohen N, Goldberg MB, Filbin MR, Li JZ. SARS-CoV-2 viremia is associated with distinct proteomic pathways and predicts COVID-19 outcomes. J Clin Invest 2021; 131:148635. [PMID: 34196300 PMCID: PMC8245177 DOI: 10.1172/jci148635] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUNDSARS-CoV-2 plasma viremia has been associated with severe disease and death in COVID-19 in small-scale cohort studies. The mechanisms behind this association remain elusive.METHODSWe evaluated the relationship between SARS-CoV-2 viremia, disease outcome, and inflammatory and proteomic profiles in a cohort of COVID-19 emergency department participants. SARS-CoV-2 viral load was measured using a quantitative reverse transcription PCR-based platform. Proteomic data were generated with Proximity Extension Assay using the Olink platform.RESULTSThis study included 300 participants with nucleic acid test-confirmed COVID-19. Plasma SARS-CoV-2 viremia levels at the time of presentation predicted adverse disease outcomes, with an adjusted OR of 10.6 (95% CI 4.4-25.5, P < 0.001) for severe disease (mechanical ventilation and/or 28-day mortality) and 3.9 (95% CI 1.5-10.1, P = 0.006) for 28-day mortality. Proteomic analyses revealed prominent proteomic pathways associated with SARS-CoV-2 viremia, including upregulation of SARS-CoV-2 entry factors (ACE2, CTSL, FURIN), heightened markers of tissue damage to the lungs, gastrointestinal tract, and endothelium/vasculature, and alterations in coagulation pathways.CONCLUSIONThese results highlight the cascade of vascular and tissue damage associated with SARS-CoV-2 plasma viremia that underlies its ability to predict COVID-19 disease outcomes.FUNDINGMark and Lisa Schwartz; the National Institutes of Health (U19AI082630); the American Lung Association; the Executive Committee on Research at Massachusetts General Hospital; the Chan Zuckerberg Initiative; Arthur, Sandra, and Sarah Irving for the David P. Ryan, MD, Endowed Chair in Cancer Research; an EMBO Long-Term Fellowship (ALTF 486-2018); a Cancer Research Institute/Bristol Myers Squibb Fellowship (CRI2993); the Harvard Catalyst/Harvard Clinical and Translational Science Center (National Center for Advancing Translational Sciences, NIH awards UL1TR001102 and UL1TR002541-01); and by the Harvard University Center for AIDS Research (National Institute of Allergy and Infectious Diseases, 5P30AI060354).
Collapse
Affiliation(s)
- Yijia Li
- Brigham and Women’s Hospital and
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexis M. Schneider
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Arnav Mehta
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Center for Cancer Research, Department of Medicine, and
| | - Moshe Sade-Feldman
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Cancer Research, Department of Medicine, and
| | - Kyle R. Kays
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Matteo Gentili
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Nicole C. Charland
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anna L.K. Gonye
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Cancer Research, Department of Medicine, and
| | - Irena Gushterova
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Cancer Research, Department of Medicine, and
| | - Hargun K. Khanna
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas J. LaSalle
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Cancer Research, Department of Medicine, and
| | | | - Brendan M. Lilley
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carl L. Lodenstein
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kasidet Manakongtreecheep
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Justin D. Margolin
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brenna N. McKaig
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Blair A. Parry
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Maricarmen Rojas-Lopez
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Brian C. Russo
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Nihaarika Sharma
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Jessica Tantivit
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Molly F. Thomas
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | - Alexandra-Chloé Villani
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Nir Hacohen
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Cancer Research, Department of Medicine, and
| | - Marcia B. Goldberg
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael R. Filbin
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | |
Collapse
|
331
|
Shukla N, Roelle SM, Suzart VG, Bruchez AM, Matreyek KA. Mutants of human ACE2 differentially promote SARS-CoV and SARS-CoV-2 spike mediated infection. PLoS Pathog 2021; 17:e1009715. [PMID: 34270613 PMCID: PMC8284657 DOI: 10.1371/journal.ppat.1009715] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/15/2021] [Indexed: 01/10/2023] Open
Abstract
SARS-CoV and SARS-CoV-2 encode spike proteins that bind human ACE2 on the cell surface to enter target cells during infection. A small fraction of humans encode variants of ACE2, thus altering the biochemical properties at the protein interaction interface. These and other ACE2 coding mutants can reveal how the spike proteins of each virus may differentially engage the ACE2 protein surface during infection. We created an engineered HEK 293T cell line for facile stable transgenic modification, and expressed the major human ACE2 allele or 28 of its missense mutants, 24 of which are possible through single nucleotide changes from the human reference sequence. Infection with SARS-CoV or SARS-CoV-2 spike pseudotyped lentiviruses revealed that high ACE2 cell-surface expression could mask the effects of impaired binding during infection. Drastically reducing ACE2 cell surface expression revealed a range of infection efficiencies across the panel of mutants. Our infection results revealed a non-linear relationship between soluble SARS-CoV-2 RBD binding to ACE2 and pseudovirus infection, supporting a major role for binding avidity during entry. While ACE2 mutants D355N, R357A, and R357T abrogated entry by both SARS-CoV and SARS-CoV-2 spike proteins, the Y41A mutant inhibited SARS-CoV entry much more than SARS-CoV-2, suggesting differential utilization of the ACE2 side-chains within the largely overlapping interaction surfaces utilized by the two CoV spike proteins. These effects correlated well with cytopathic effects observed during SARS-CoV-2 replication in ACE2-mutant cells. The panel of ACE2 mutants also revealed altered ACE2 surface dependencies by the N501Y spike variant, including a near-complete utilization of the K353D ACE2 variant, despite decreased infection mediated by the parental SARS-CoV-2 spike. Our results clarify the relationship between ACE2 abundance, binding, and infection, for various SARS-like coronavirus spike proteins and their mutants, and inform our understanding for how changes to ACE2 sequence may correspond with different susceptibilities to infection.
Collapse
Affiliation(s)
- Nidhi Shukla
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Sarah M. Roelle
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Vinicius G. Suzart
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Anna M. Bruchez
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Kenneth A. Matreyek
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| |
Collapse
|
332
|
Wang Z, Muecksch F, Schaefer-Babajew D, Finkin S, Viant C, Gaebler C, Hoffmann HH, Barnes CO, Cipolla M, Ramos V, Oliveira TY, Cho A, Schmidt F, Da Silva J, Bednarski E, Aguado L, Yee J, Daga M, Turroja M, Millard KG, Jankovic M, Gazumyan A, Zhao Z, Rice CM, Bieniasz PD, Caskey M, Hatziioannou T, Nussenzweig MC. Naturally enhanced neutralizing breadth against SARS-CoV-2 one year after infection. Nature 2021; 595:426-431. [PMID: 34126625 PMCID: PMC8277577 DOI: 10.1038/s41586-021-03696-9] [Citation(s) in RCA: 540] [Impact Index Per Article: 135.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 06/04/2021] [Indexed: 02/05/2023]
Abstract
More than one year after its inception, the coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains difficult to control despite the availability of several working vaccines. Progress in controlling the pandemic is slowed by the emergence of variants that appear to be more transmissible and more resistant to antibodies1,2. Here we report on a cohort of 63 individuals who have recovered from COVID-19 assessed at 1.3, 6.2 and 12 months after SARS-CoV-2 infection, 41% of whom also received mRNA vaccines3,4. In the absence of vaccination, antibody reactivity to the receptor binding domain (RBD) of SARS-CoV-2, neutralizing activity and the number of RBD-specific memory B cells remain relatively stable between 6 and 12 months after infection. Vaccination increases all components of the humoral response and, as expected, results in serum neutralizing activities against variants of concern similar to or greater than the neutralizing activity against the original Wuhan Hu-1 strain achieved by vaccination of naive individuals2,5-8. The mechanism underlying these broad-based responses involves ongoing antibody somatic mutation, memory B cell clonal turnover and development of monoclonal antibodies that are exceptionally resistant to SARS-CoV-2 RBD mutations, including those found in the variants of concern4,9. In addition, B cell clones expressing broad and potent antibodies are selectively retained in the repertoire over time and expand markedly after vaccination. The data suggest that immunity in convalescent individuals will be very long lasting and that convalescent individuals who receive available mRNA vaccines will produce antibodies and memory B cells that should be protective against circulating SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Zijun Wang
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Frauke Muecksch
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | | | - Shlomo Finkin
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Charlotte Viant
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Christian Gaebler
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Hans- Heinrich Hoffmann
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Christopher O Barnes
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Melissa Cipolla
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Victor Ramos
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Alice Cho
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Fabian Schmidt
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | - Justin Da Silva
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | - Eva Bednarski
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | - Lauren Aguado
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Jim Yee
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Mridushi Daga
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Martina Turroja
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Katrina G Millard
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Mila Jankovic
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Anna Gazumyan
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
| | - Zhen Zhao
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Paul D Bieniasz
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
| | - Marina Caskey
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA.
| | | | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
| |
Collapse
|
333
|
Identification of a High-Frequency Intrahost SARS-CoV-2 Spike Variant with Enhanced Cytopathic and Fusogenic Effects. mBio 2021; 12:e0078821. [PMID: 34182784 PMCID: PMC8262852 DOI: 10.1128/mbio.00788-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a virus that is continuously evolving. Although its RNA-dependent RNA polymerase exhibits some exonuclease proofreading activity, viral sequence diversity can be produced by replication errors and host factors. A diversity of genetic variants can be observed in the intrahost viral population structure of infected individuals. Most mutations will follow a neutral molecular evolution and will not make significant contributions to variations within and between infected hosts. Herein, we profiled the intrasample genetic diversity of SARS-CoV-2 variants, also known as quasispecies, using high-throughput sequencing data sets from 15,289 infected individuals and infected cell lines. Despite high mutational background, we identified recurrent intragenetic variable positions in the samples analyzed, including several positions at the end of the gene encoding the viral spike (S) protein. Strikingly, we observed a high frequency of C→A missense mutations resulting in the S protein lacking the last 20 amino acids (SΔ20). We found that this truncated S protein undergoes increased processing and increased syncytium formation, presumably due to escaping M protein retention in intracellular compartments. Our findings suggest the emergence of a high-frequency viral sublineage that is not horizontally transmitted but potentially involved in intrahost disease cytopathic effects. IMPORTANCE The mutation rate and evolution of RNA viruses correlate with viral adaptation. While most mutations do not make significant contributions to viral molecular evolution, some are naturally selected and produce variants through positive selection. Many SARS-CoV-2 variants have been recently described and show phenotypic selection toward more infectious viruses. Our study describes another type of variant that does not contribute to interhost heterogeneity but rather phenotypic selection toward variants that might have increased cytopathic effects. We identified that a C-terminal truncation of the spike protein removes an important endoplasmic reticulum (ER) retention signal, which consequently results in a spike variant that easily travels through the Golgi complex toward the plasma membrane in a preactivated conformation, leading to increased syncytium formation.
Collapse
|
334
|
Meng B, Kemp SA, Papa G, Datir R, Ferreira IATM, Marelli S, Harvey WT, Lytras S, Mohamed A, Gallo G, Thakur N, Collier DA, Mlcochova P, Duncan LM, Carabelli AM, Kenyon JC, Lever AM, De Marco A, Saliba C, Culap K, Cameroni E, Matheson NJ, Piccoli L, Corti D, James LC, Robertson DL, Bailey D, Gupta RK. Recurrent emergence of SARS-CoV-2 spike deletion H69/V70 and its role in the Alpha variant B.1.1.7. Cell Rep 2021. [PMID: 34166617 DOI: 10.1101/2020.12.14.422555] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
We report severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike ΔH69/V70 in multiple independent lineages, often occurring after acquisition of receptor binding motif replacements such as N439K and Y453F, known to increase binding affinity to the ACE2 receptor and confer antibody escape. In vitro, we show that, although ΔH69/V70 itself is not an antibody evasion mechanism, it increases infectivity associated with enhanced incorporation of cleaved spike into virions. ΔH69/V70 is able to partially rescue infectivity of spike proteins that have acquired N439K and Y453F escape mutations by increased spike incorporation. In addition, replacement of the H69 and V70 residues in the Alpha variant B.1.1.7 spike (where ΔH69/V70 occurs naturally) impairs spike incorporation and entry efficiency of the B.1.1.7 spike pseudotyped virus. Alpha variant B.1.1.7 spike mediates faster kinetics of cell-cell fusion than wild-type Wuhan-1 D614G, dependent on ΔH69/V70. Therefore, as ΔH69/V70 compensates for immune escape mutations that impair infectivity, continued surveillance for deletions with functional effects is warranted.
Collapse
Affiliation(s)
- Bo Meng
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Steven A Kemp
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; Division of Infection and Immunity, University College London, London, UK
| | - Guido Papa
- MRC - Laboratory of Molecular Biology, Cambridge, UK
| | - Rawlings Datir
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Isabella A T M Ferreira
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Sara Marelli
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - William T Harvey
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK; MRC - University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Spyros Lytras
- MRC - University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | | | | | - Dami A Collier
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; Division of Infection and Immunity, University College London, London, UK
| | - Petra Mlcochova
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lidia M Duncan
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Julia C Kenyon
- Department of Medicine, University of Cambridge, Cambridge, UK; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Andrew M Lever
- Department of Medicine, University of Cambridge, Cambridge, UK; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anna De Marco
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Christian Saliba
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Katja Culap
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Elisabetta Cameroni
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Nicholas J Matheson
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; NHS Blood and Transplant, Cambridge, UK
| | - Luca Piccoli
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Leo C James
- MRC - Laboratory of Molecular Biology, Cambridge, UK
| | | | | | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; Africa Health Research Institute, Durban, South Africa.
| |
Collapse
|
335
|
Meng B, Kemp SA, Papa G, Datir R, Ferreira IATM, Marelli S, Harvey WT, Lytras S, Mohamed A, Gallo G, Thakur N, Collier DA, Mlcochova P, Duncan LM, Carabelli AM, Kenyon JC, Lever AM, De Marco A, Saliba C, Culap K, Cameroni E, Matheson NJ, Piccoli L, Corti D, James LC, Robertson DL, Bailey D, Gupta RK. Recurrent emergence of SARS-CoV-2 spike deletion H69/V70 and its role in the Alpha variant B.1.1.7. Cell Rep 2021; 35:109292. [PMID: 34166617 PMCID: PMC8185188 DOI: 10.1016/j.celrep.2021.109292] [Citation(s) in RCA: 318] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/29/2021] [Accepted: 06/02/2021] [Indexed: 12/23/2022] Open
Abstract
We report severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike ΔH69/V70 in multiple independent lineages, often occurring after acquisition of receptor binding motif replacements such as N439K and Y453F, known to increase binding affinity to the ACE2 receptor and confer antibody escape. In vitro, we show that, although ΔH69/V70 itself is not an antibody evasion mechanism, it increases infectivity associated with enhanced incorporation of cleaved spike into virions. ΔH69/V70 is able to partially rescue infectivity of spike proteins that have acquired N439K and Y453F escape mutations by increased spike incorporation. In addition, replacement of the H69 and V70 residues in the Alpha variant B.1.1.7 spike (where ΔH69/V70 occurs naturally) impairs spike incorporation and entry efficiency of the B.1.1.7 spike pseudotyped virus. Alpha variant B.1.1.7 spike mediates faster kinetics of cell-cell fusion than wild-type Wuhan-1 D614G, dependent on ΔH69/V70. Therefore, as ΔH69/V70 compensates for immune escape mutations that impair infectivity, continued surveillance for deletions with functional effects is warranted.
Collapse
Affiliation(s)
- Bo Meng
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Steven A Kemp
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; Division of Infection and Immunity, University College London, London, UK
| | - Guido Papa
- MRC - Laboratory of Molecular Biology, Cambridge, UK
| | - Rawlings Datir
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Isabella A T M Ferreira
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Sara Marelli
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - William T Harvey
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK; MRC - University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Spyros Lytras
- MRC - University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | | | | | - Dami A Collier
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; Division of Infection and Immunity, University College London, London, UK
| | - Petra Mlcochova
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lidia M Duncan
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Julia C Kenyon
- Department of Medicine, University of Cambridge, Cambridge, UK; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Andrew M Lever
- Department of Medicine, University of Cambridge, Cambridge, UK; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anna De Marco
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Christian Saliba
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Katja Culap
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Elisabetta Cameroni
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Nicholas J Matheson
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; NHS Blood and Transplant, Cambridge, UK
| | - Luca Piccoli
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Leo C James
- MRC - Laboratory of Molecular Biology, Cambridge, UK
| | | | | | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; Africa Health Research Institute, Durban, South Africa.
| |
Collapse
|
336
|
Tada T, Dcosta BM, Samanovic MI, Herati RS, Cornelius A, Zhou H, Vaill A, Kazmierski W, Mulligan MJ, Landau NR. Convalescent-Phase Sera and Vaccine-Elicited Antibodies Largely Maintain Neutralizing Titer against Global SARS-CoV-2 Variant Spikes. mBio 2021; 12:e0069621. [PMID: 34060334 PMCID: PMC8262901 DOI: 10.1128/mbio.00696-21] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/28/2021] [Indexed: 12/21/2022] Open
Abstract
The increasing prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with spike protein mutations raises concerns that antibodies elicited by natural infection or vaccination and therapeutic monoclonal antibodies will become less effective. We show that convalescent-phase sera neutralize pseudotyped viruses with the B.1.1.7, B.1.351, B.1.1.248, COH.20G/677H, 20A.EU2, and mink cluster 5 spike proteins with only a minor loss in titer. Similarly, antibodies elicited by Pfizer BNT162b2 vaccination neutralized B.1.351 and B.1.1.248 with only a 3-fold decrease in titer, an effect attributable to E484K. Analysis of the Regeneron monoclonal antibodies REGN10933 and REGN10987 showed that REGN10933 has lost neutralizing activity against the B.1.351 and B.1.1.248 pseudotyped viruses, and the cocktail is 9- to 15-fold decreased in titer. These findings suggest that antibodies elicited by natural infection and by the Pfizer vaccine will maintain protection against the B.1.1.7, B.1.351, and B.1.1.248 variants but that monoclonal antibody therapy may be less effective for patients infected with B.1.351 or B.1.1.248 SARS-CoV-2. IMPORTANCE The rapid evolution of SARS-CoV-2 variants has raised concerns with regard to their potential to escape from vaccine-elicited antibodies and anti-spike protein monoclonal antibodies. We report here on an analysis of sera from recovered patients and vaccinated individuals and on neutralization by Regeneron therapeutic monoclonal antibodies. Overall, the variants were neutralized nearly as well as the wild-type pseudotyped virus. The B.1.351 variant was somewhat resistant to vaccine-elicited antibodies but was still readily neutralized. One of the two Regeneron therapeutic monoclonal antibodies seems to have lost most of its activity against the B.1.351 variant, raising concerns that the combination therapy might be less effective for some patients. The findings should alleviate concerns that vaccines will become ineffective but suggest the importance of continued surveillance for potential new variants.
Collapse
Affiliation(s)
- Takuya Tada
- Department of Microbiology, NYU Grossman School of Medicine, New York, New York, USA
| | - Belinda M. Dcosta
- Department of Microbiology, NYU Grossman School of Medicine, New York, New York, USA
| | - Marie I. Samanovic
- NYU Langone Vaccine Center, NYU Grossman School of Medicine, New York, New York, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Ramin S. Herati
- NYU Langone Vaccine Center, NYU Grossman School of Medicine, New York, New York, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Amber Cornelius
- NYU Langone Vaccine Center, NYU Grossman School of Medicine, New York, New York, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Hao Zhou
- Department of Microbiology, NYU Grossman School of Medicine, New York, New York, USA
| | - Ada Vaill
- Biohaven Pharmaceuticals, Inc., New Haven, Connecticut, USA
| | - Wes Kazmierski
- Biohaven Pharmaceuticals, Inc., New Haven, Connecticut, USA
| | - Mark J. Mulligan
- NYU Langone Vaccine Center, NYU Grossman School of Medicine, New York, New York, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Nathaniel R. Landau
- Department of Microbiology, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
337
|
SARS-CoV-2 variant B.1.617 is resistant to bamlanivimab and evades antibodies induced by infection and vaccination. Cell Rep 2021; 36:109415. [PMID: 34270919 PMCID: PMC8238662 DOI: 10.1016/j.celrep.2021.109415] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/28/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants threatens efforts to contain the coronavirus disease 2019 (COVID-19) pandemic. The number of COVID-19 cases and deaths in India has risen steeply, and a SARS-CoV-2 variant, B.1.617, is believed to be responsible for many of these cases. The spike protein of B.1.617 harbors two mutations in the receptor binding domain, which interacts with the angiotensin converting enzyme 2 (ACE2) receptor and constitutes the main target of neutralizing antibodies. Therefore, we analyze whether B.1.617 is more adept in entering cells and/or evades antibody responses. B.1.617 enters two of eight cell lines tested with roughly 50% increased efficiency and is equally inhibited by two entry inhibitors. In contrast, B.1.617 is resistant against bamlanivimab, an antibody used for COVID-19 treatment. B.1.617 evades antibodies induced by infection or vaccination, although less so than the B.1.351 variant. Collectively, our study reveals that antibody evasion of B.1.617 may contribute to the rapid spread of this variant.
Collapse
|
338
|
ACE2 interaction with cytoplasmic PDZ protein enhances SARS-CoV-2 invasion. iScience 2021; 24:102770. [PMID: 34189428 PMCID: PMC8223119 DOI: 10.1016/j.isci.2021.102770] [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: 03/23/2021] [Revised: 05/23/2021] [Accepted: 06/21/2021] [Indexed: 12/26/2022] Open
Abstract
SARS-CoV-2 is responsible for the global COVID-19 pandemic. Angiotensin converting enzyme 2 (ACE2) is the membrane-delimited receptor for SARS-CoV-2. Lung, intestine, and kidney, major sites of viral infection, express ACE2 that harbors an intracellular, carboxy-terminal PDZ-recognition motif. These organs prominently express the PDZ protein Na+/H+ exchanger regulatory factor-1 (NHERF1). Here, we report NHERF1 tethers ACE2 and augments SARS-CoV-2 cell entry. ACE2 directly binds both NHERF1 PDZ domains. Disruption of either NHERF1 PDZ core-binding motif or the ACE2 PDZ recognition sequence eliminates interaction. Proximity ligation assays establish that ACE2 and NHERF1 interact at constitutive expression levels in human lung and intestine cells. Ablating ACE2 interaction with NHERF1 accelerated SARS-CoV-2 cell entry. Conversely, elimination of the ACE2 C-terminal PDZ-binding motif decreased ACE2 membrane residence and reduced pseudotyped virus entry. We conclude that the PDZ interaction of ACE2 with NHERF1 facilitates SARS-CoV-2 internalization. β-Arrestin is likely indispensable, as with G protein-coupled receptors. The SARS-CoV-2 receptor ACE2 C-terminal PDZ-recognition motif 802QTSF805 binds to NHERF1 NHERF1 and ACE2 interact directly in SARS-CoV-2-susceptible lung and intestine cells NHERF1 expression correlates with SARS-CoV-2 entry by regulating ACE2 membrane abundance β-Arrestins may cooperate with NHERF1 to promote ACE2-mediated SARS-CoV-2 cell entry
Collapse
|
339
|
Systematic analysis of SARS-CoV-2 infection of an ACE2-negative human airway cell. Cell Rep 2021; 36:109364. [PMID: 34214467 PMCID: PMC8220945 DOI: 10.1016/j.celrep.2021.109364] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/25/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) variants govern transmissibility, responsiveness to vaccination, and disease severity. In a screen for new models of SARS-CoV-2 infection, we identify human H522 lung adenocarcinoma cells as naturally permissive to SARS-CoV-2 infection despite complete absence of angiotensin-converting enzyme 2 (ACE2) expression. Remarkably, H522 infection requires the E484D S variant; viruses expressing wild-type S are not infectious. Anti-S monoclonal antibodies differentially neutralize SARS-CoV-2 E484D S in H522 cells as compared to ACE2-expressing cells. Sera from vaccinated individuals block this alternative entry mechanism, whereas convalescent sera are less effective. Although the H522 receptor remains unknown, depletion of surface heparan sulfates block H522 infection. Temporally resolved transcriptomic and proteomic profiling reveal alterations in cell cycle and the antiviral host cell response, including MDA5-dependent activation of type I interferon signaling. These findings establish an alternative SARS-CoV-2 host cell receptor for the E484D SARS-CoV-2 variant, which may impact tropism of SARS-CoV-2 and consequently human disease pathogenesis.
Collapse
|
340
|
Safiabadi Tali SH, LeBlanc JJ, Sadiq Z, Oyewunmi OD, Camargo C, Nikpour B, Armanfard N, Sagan SM, Jahanshahi-Anbuhi S. Tools and Techniques for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2)/COVID-19 Detection. Clin Microbiol Rev 2021; 34:e00228-20. [PMID: 33980687 PMCID: PMC8142517 DOI: 10.1128/cmr.00228-20] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory disease coronavirus 2 (SARS-CoV-2), has led to millions of confirmed cases and deaths worldwide. Efficient diagnostic tools are in high demand, as rapid and large-scale testing plays a pivotal role in patient management and decelerating disease spread. This paper reviews current technologies used to detect SARS-CoV-2 in clinical laboratories as well as advances made for molecular, antigen-based, and immunological point-of-care testing, including recent developments in sensor and biosensor devices. The importance of the timing and type of specimen collection is discussed, along with factors such as disease prevalence, setting, and methods. Details of the mechanisms of action of the various methodologies are presented, along with their application span and known performance characteristics. Diagnostic imaging techniques and biomarkers are also covered, with an emphasis on their use for assessing COVID-19 or monitoring disease severity or complications. While the SARS-CoV-2 literature is rapidly evolving, this review highlights topics of interest that have occurred during the pandemic and the lessons learned throughout. Exploring a broad armamentarium of techniques for detecting SARS-CoV-2 will ensure continued diagnostic support for clinicians, public health, and infection prevention and control for this pandemic and provide advice for future pandemic preparedness.
Collapse
Affiliation(s)
- Seyed Hamid Safiabadi Tali
- Department of Chemical and Materials Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
- Department of Mechanical, Industrial, and Aerospace Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
| | - Jason J LeBlanc
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medicine (Infectious Diseases), Dalhousie University, Halifax, Nova Scotia, Canada
- Division of Microbiology, Department of Pathology and Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
| | - Zubi Sadiq
- Department of Chemical and Materials Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
| | - Oyejide Damilola Oyewunmi
- Department of Chemical and Materials Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
| | - Carolina Camargo
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
| | - Bahareh Nikpour
- Department of Electrical and Computer Engineering, McGill University, Montréal, Québec, Canada
| | - Narges Armanfard
- Department of Electrical and Computer Engineering, McGill University, Montréal, Québec, Canada
- Mila-Quebec AI Institute, Montréal, Québec, Canada
| | - Selena M Sagan
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Sana Jahanshahi-Anbuhi
- Department of Chemical and Materials Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
| |
Collapse
|
341
|
Abstract
The coronavirus disease 2019 (COVID‐19) pandemic has triggered a global health emergency and brought disaster to humans. Tremendous efforts have been made to control the pandemic, among which neutralizing antibodies (NAbs) are of specific interest to researchers. Neutralizing antibodies are generated within weeks after infection or immunization and can protect cells from virus intrusion and confer protective immunity to cells. Thus, production of NAbs is considered as a main goal for severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) vaccines and NAbs may be used for patient treatment in the form of monoclonal antibodies. Neutralization assays are capable of quantitatively detecting NAbs against SARS‐CoV‐2, allowing to explore the relationship between the level of NAbs and the severity of the disease, and may predict the possibility of re‐infection in COVID‐19 patients. They can also be used to test the effects of monoclonal antibodies, convalescent plasma and vaccines. At present, wild‐type virus neutralization assay remains the gold standard for measuring Nabs, while pseudovirus neutralization assays, Surrogate virus neutralization test (sVNT) and high‐throughput versions of neutralization assays are popular alternatives with their own advantages and disadvantages. In this review article, we summarize the characteristics and recent progress of SARS‐CoV‐2 neutralization assays. Special attention is given to the current limitations of various neutralization assays so as to promote new possible strategies with NAbs by which rapid SARS‐CoV‐2 serological diagnosis and antiviral screening in the future will be achieved.
Collapse
Affiliation(s)
- Yuying Lu
- Department of Epidemiology School of Public Health Sun Yat‐Sen University Guangzhou China
| | - Jin Wang
- Department of Epidemiology School of Public Health Sun Yat‐Sen University Guangzhou China
| | - Qianlin Li
- Department of Epidemiology School of Public Health Sun Yat‐Sen University Guangzhou China
| | - Huan Hu
- Department of Epidemiology School of Public Health Sun Yat‐Sen University Guangzhou China
| | - Jiahai Lu
- Department of Epidemiology School of Public Health Sun Yat‐Sen University Guangzhou China
| | - Zeliang Chen
- Department of Epidemiology School of Public Health Sun Yat‐Sen University Guangzhou China
| |
Collapse
|
342
|
Kruglova N, Siniavin A, Gushchin V, Mazurov D. Different Neutralization Sensitivity of SARS-CoV-2 Cell-to-Cell and Cell-Free Modes of Infection to Convalescent Sera. Viruses 2021; 13:1133. [PMID: 34204732 PMCID: PMC8231521 DOI: 10.3390/v13061133] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/01/2021] [Accepted: 06/09/2021] [Indexed: 12/14/2022] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has posed a global threat to human lives and economics. One of the best ways to determine protection against the infection is to quantify the neutralizing activity of serum antibodies. Multiple assays have been developed to validate SARS-CoV-2 neutralization; most of them utilized lentiviral or vesicular stomatitis virus-based particles pseudotyped with the spike (S) protein, making them safe and acceptable to work with in many labs. However, these systems are only capable of measuring infection with purified particles. This study has developed a pseudoviral assay with replication-dependent reporter vectors that can accurately quantify the level of infection directly from the virus producing cell to the permissive target cell. Comparative analysis of cell-free and cell-to-cell infection revealed that the neutralizing activity of convalescent sera was more than tenfold lower in cell cocultures than in the cell-free mode of infection. As the pseudoviral system could not properly model the mechanisms of SARS-CoV-2 transmission, similar experiments were performed with replication-competent coronavirus, which detected nearly complete SARS-CoV-2 cell-to-cell infection resistance to neutralization by convalescent sera. These findings suggest that the cell-to-cell mode of SARS-CoV-2 transmission, for which the mechanisms are largely unknown, could be of great importance for treatment and prevention of COVID-19.
Collapse
Affiliation(s)
- Natalia Kruglova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology RAS, 119334 Moscow, Russia;
| | - Andrei Siniavin
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (A.S.); (V.G.)
- Department of Molecular Neuroimmune Signalling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Vladimir Gushchin
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (A.S.); (V.G.)
| | - Dmitriy Mazurov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology RAS, 119334 Moscow, Russia;
| |
Collapse
|
343
|
Scheid JF, Barnes CO, Eraslan B, Hudak A, Keeffe JR, Cosimi LA, Brown EM, Muecksch F, Weisblum Y, Zhang S, Delorey T, Woolley AE, Ghantous F, Park SM, Phillips D, Tusi B, Huey-Tubman KE, Cohen AA, Gnanapragasam PNP, Rzasa K, Hatziioanno T, Durney MA, Gu X, Tada T, Landau NR, West AP, Rozenblatt-Rosen O, Seaman MS, Baden LR, Graham DB, Deguine J, Bieniasz PD, Regev A, Hung D, Bjorkman PJ, Xavier RJ. B cell genomics behind cross-neutralization of SARS-CoV-2 variants and SARS-CoV. Cell 2021; 184:3205-3221.e24. [PMID: 34015271 PMCID: PMC8064835 DOI: 10.1016/j.cell.2021.04.032] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/26/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023]
Abstract
Monoclonal antibodies (mAbs) are a focus in vaccine and therapeutic design to counteract severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants. Here, we combined B cell sorting with single-cell VDJ and RNA sequencing (RNA-seq) and mAb structures to characterize B cell responses against SARS-CoV-2. We show that the SARS-CoV-2-specific B cell repertoire consists of transcriptionally distinct B cell populations with cells producing potently neutralizing antibodies (nAbs) localized in two clusters that resemble memory and activated B cells. Cryo-electron microscopy structures of selected nAbs from these two clusters complexed with SARS-CoV-2 spike trimers show recognition of various receptor-binding domain (RBD) epitopes. One of these mAbs, BG10-19, locks the spike trimer in a closed conformation to potently neutralize SARS-CoV-2, the recently arising mutants B.1.1.7 and B.1.351, and SARS-CoV and cross-reacts with heterologous RBDs. Together, our results characterize transcriptional differences among SARS-CoV-2-specific B cells and uncover cross-neutralizing Ab targets that will inform immunogen and therapeutic design against coronaviruses.
Collapse
MESH Headings
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/chemistry
- Antibodies, Viral/immunology
- Antigen-Antibody Complex/chemistry
- Antigen-Antibody Complex/metabolism
- Antigen-Antibody Reactions
- B-Lymphocytes/cytology
- B-Lymphocytes/metabolism
- B-Lymphocytes/virology
- COVID-19/pathology
- COVID-19/virology
- Cryoelectron Microscopy
- Crystallography, X-Ray
- Gene Expression Profiling
- Humans
- Immunoglobulin A/immunology
- Immunoglobulin Variable Region/chemistry
- Immunoglobulin Variable Region/genetics
- Protein Domains/immunology
- Protein Multimerization
- Protein Structure, Quaternary
- SARS-CoV-2/immunology
- SARS-CoV-2/isolation & purification
- SARS-CoV-2/metabolism
- Sequence Analysis, RNA
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
Collapse
Affiliation(s)
- Johannes F Scheid
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Christopher O Barnes
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Basak Eraslan
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Andrew Hudak
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Jennifer R Keeffe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Lisa A Cosimi
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Eric M Brown
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Frauke Muecksch
- Laboratory of Molecular Virology, The Rockefeller University, New York, NY 10065, USA
| | - Yiska Weisblum
- Laboratory of Molecular Virology, The Rockefeller University, New York, NY 10065, USA
| | - Shuting Zhang
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Toni Delorey
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ann E Woolley
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Fadi Ghantous
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Sung-Moo Park
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Devan Phillips
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Betsabeh Tusi
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Kathryn E Huey-Tubman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Alexander A Cohen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | - Kara Rzasa
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Theodora Hatziioanno
- Laboratory of Molecular Virology, The Rockefeller University, New York, NY 10065, USA
| | - Michael A Durney
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Xiebin Gu
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Takuya Tada
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Nathaniel R Landau
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Anthony P West
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Orit Rozenblatt-Rosen
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael S Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Lindsey R Baden
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel B Graham
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jacques Deguine
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Paul D Bieniasz
- Laboratory of Molecular Virology, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Deborah Hung
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Ramnik J Xavier
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
344
|
Hacisuleyman E, Hale C, Saito Y, Blachere NE, Bergh M, Conlon EG, Schaefer-Babajew DJ, DaSilva J, Muecksch F, Gaebler C, Lifton R, Nussenzweig MC, Hatziioannou T, Bieniasz PD, Darnell RB. Vaccine Breakthrough Infections with SARS-CoV-2 Variants. N Engl J Med 2021; 384:2212-2218. [PMID: 33882219 PMCID: PMC8117968 DOI: 10.1056/nejmoa2105000] [Citation(s) in RCA: 518] [Impact Index Per Article: 129.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Emerging variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are of clinical concern. In a cohort of 417 persons who had received the second dose of BNT162b2 (Pfizer-BioNTech) or mRNA-1273 (Moderna) vaccine at least 2 weeks previously, we identified 2 women with vaccine breakthrough infection. Despite evidence of vaccine efficacy in both women, symptoms of coronavirus disease 2019 developed, and they tested positive for SARS-CoV-2 by polymerase-chain-reaction testing. Viral sequencing revealed variants of likely clinical importance, including E484K in 1 woman and three mutations (T95I, del142-144, and D614G) in both. These observations indicate a potential risk of illness after successful vaccination and subsequent infection with variant virus, and they provide support for continued efforts to prevent and diagnose infection and to characterize variants in vaccinated persons. (Funded by the National Institutes of Health and others.).
Collapse
Affiliation(s)
- Ezgi Hacisuleyman
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Caryn Hale
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Yuhki Saito
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Nathalie E Blachere
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Marissa Bergh
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Erin G Conlon
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Dennis J Schaefer-Babajew
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Justin DaSilva
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Frauke Muecksch
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Christian Gaebler
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Richard Lifton
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Michel C Nussenzweig
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Theodora Hatziioannou
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Paul D Bieniasz
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| | - Robert B Darnell
- From the Laboratory of Molecular Neuro-oncology (E.H., C.H., Y.S., N.E.B., M.B., E.G.C., R.B.D.), the Laboratory of Molecular Immunology (D.J.S.-B., C.G., M.C.N.), the Laboratory of Human Genetics and Genomics (R.L.), the Laboratory of Retrovirology (J.D., F.M., T.H., P.D.B.), and the Howard Hughes Medical Institute (M.C.N., P.D.B., R.B.D.), Rockefeller University, New York
| |
Collapse
|
345
|
Scaglione A, Opp S, Hurtado A, Lin Z, Pampeno C, Noval MG, Thannickal SA, Stapleford KA, Meruelo D. Combination of a Sindbis-SARS-CoV-2 spike vaccine and αOX40 antibody elicits protective immunity against SARS-CoV-2 induced disease and potentiates long-term SARS-CoV-2-specific humoral and T-cell immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.05.28.446009. [PMID: 34075383 PMCID: PMC8168399 DOI: 10.1101/2021.05.28.446009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The COVID-19 pandemic caused by the coronavirus SARS-CoV-2 is a major global public threat. Currently, a worldwide effort has been mounted to generate billions of effective SARS-CoV-2 vaccine doses to immunize the world's population at record speeds. However, there is still demand for alternative effective vaccines that rapidly confer long-term protection and rely upon cost-effective, easily scaled-up manufacturing. Here, we present a Sindbis alphavirus vector (SV), transiently expressing the SARS-CoV-2 spike protein (SV.Spike), combined with the OX40 immunostimulatory antibody (αOX40) as a novel, highly effective vaccine approach. We show that SV.Spike plus αOX40 elicits long-lasting neutralizing antibodies and a vigorous T-cell response in mice. Protein binding, immunohistochemical and cellular infection assays all show that vaccinated mice sera inhibits spike functions. Immunophenotyping, RNA Seq transcriptome profiles and metabolic analysis indicate a reprogramming of T-cells in vaccinated mice. Activated T-cells were found to mobilize to lung tissue. Most importantly, SV.Spike plus αOX40 provided robust immune protection against infection with authentic coronavirus in transgenic mice expressing the human ACE2 receptor (hACE2-Tg). Finally, our immunization strategy induced strong effector memory response, potentiating protective immunity against re-exposure to SARS-CoV-2 spike protein. Our results show the potential of a new Sindbis virus-based vaccine platform to counteract waning immune response that can be used as a new candidate to combat SARS-CoV-2. Given the strong T-cell responses elicited, our vaccine is likely to be effective against variants that are proving challenging, as well as, serve as a platform to develop a broader spectrum pancoronavirus vaccine. Similarly, the vaccine approach is likely to be applicable to other pathogens.
Collapse
Affiliation(s)
- Antonella Scaglione
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Silvana Opp
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Alicia Hurtado
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ziyan Lin
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Christine Pampeno
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Maria G Noval
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sara A. Thannickal
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kenneth A. Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Daniel Meruelo
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
346
|
Wang Z, Muecksch F, Schaefer-Babajew D, Finkin S, Viant C, Gaebler C, Hoffmann HH, Barnes CO, Cipolla M, Ramos V, Oliveira TY, Cho A, Schmidt F, da Silva J, Bednarski E, Aguado L, Yee J, Daga M, Turroja M, Millard KG, Jankovic M, Gazumyan A, Zhao Z, Rice CM, Bieniasz PD, Caskey M, Hatziioannou T, Nussenzweig MC. Naturally enhanced neutralizing breadth to SARS-CoV-2 after one year. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34100013 DOI: 10.1101/2021.05.07.443175] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Over one year after its inception, the coronavirus disease-2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) remains difficult to control despite the availability of several excellent vaccines. Progress in controlling the pandemic is slowed by the emergence of variants that appear to be more transmissible and more resistant to antibodies 1,2 . Here we report on a cohort of 63 COVID-19-convalescent individuals assessed at 1.3, 6.2 and 12 months after infection, 41% of whom also received mRNA vaccines 3,4 . In the absence of vaccination antibody reactivity to the receptor binding domain (RBD) of SARS-CoV-2, neutralizing activity and the number of RBD-specific memory B cells remain relatively stable from 6 to 12 months. Vaccination increases all components of the humoral response, and as expected, results in serum neutralizing activities against variants of concern that are comparable to or greater than neutralizing activity against the original Wuhan Hu-1 achieved by vaccination of naïve individuals 2,5-8 . The mechanism underlying these broad-based responses involves ongoing antibody somatic mutation, memory B cell clonal turnover, and development of monoclonal antibodies that are exceptionally resistant to SARS-CoV-2 RBD mutations, including those found in variants of concern 4,9 . In addition, B cell clones expressing broad and potent antibodies are selectively retained in the repertoire over time and expand dramatically after vaccination. The data suggest that immunity in convalescent individuals will be very long lasting and that convalescent individuals who receive available mRNA vaccines will produce antibodies and memory B cells that should be protective against circulating SARS-CoV-2 variants.
Collapse
|
347
|
Cimolai N. Passive Immunity Should and Will Work for COVID-19 for Some Patients. Clin Hematol Int 2021; 3:47-68. [PMID: 34595467 PMCID: PMC8432400 DOI: 10.2991/chi.k.210328.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
In the absence of effective antiviral chemotherapy and still in the context of emerging vaccines for severe acute respiratory syndrome-CoV-2 infections, passive immunotherapy remains a key treatment and possible prevention strategy. What might initially be conceived as a simplified donor-recipient process, the intricacies of donor plasma, IV immunoglobulins, and monoclonal antibody modality applications are becoming more apparent. Key targets of such treatment have largely focused on virus neutralization and the specific viral components of the attachment Spike protein and its constituents (e.g., receptor binding domain, N-terminal domain). The cumulative laboratory and clinical experience suggests that beneficial protective and treatment outcomes are possible. Both a dose- and a time-dependency emerge. Lesser understood are the concepts of bioavailability and distribution. Apart from direct antigen binding from protective immunoglobulins, antibody effector functions have potential roles in outcome. In attempting to mimic the natural but variable response to infection or vaccination, a strong functional polyclonal approach attracts the potential benefits of attacking antigen diversity, high antibody avidity, antibody persistence, and protection against escape viral mutation. The availability and ease of administration for any passive immunotherapy product must be considered in the current climate of need. There is never a perfect product, but yet there is considerable room for improving patient outcomes. Given the variability of human genetics, immunity, and disease, and given the nuances of the virus and its potential for change, passive immunotherapy can be developed that will be effective for some but not all patients. An understanding of such patient variability and limitations is just as important as the understanding of the direct interactions between immunotherapy and virus.
Collapse
Affiliation(s)
- Nevio Cimolai
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, Children’s and Women’s Health Centre of British Columbia, 4480 Oak Street, Vancouver, BC, Canada V6H 3V4
| |
Collapse
|
348
|
Caniels TG, Bontjer I, van der Straten K, Poniman M, Burger JA, Appelman B, Lavell AHA, Oomen M, Godeke GJ, Valle C, Mögling R, van Willigen HDG, Wynberg E, Schinkel M, van Vught LA, Guerra D, Snitselaar JL, Chaturbhuj DN, Martin IC, Moore JP, de Jong MD, Reusken C, Sikkens JJ, Bomers MK, de Bree GJ, van Gils MJ, Eggink D, Sanders RW. Emerging SARS-CoV-2 variants of concern evade humoral immune responses from infection and vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 34100023 DOI: 10.1101/2021.05.26.21257441] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Emerging SARS-CoV-2 variants pose a threat to human immunity induced by natural infection and vaccination. We assessed the recognition of three variants of concern (B.1.1.7, B.1.351 and P.1) in cohorts of COVID-19 patients ranging in disease severity (n = 69) and recipients of the Pfizer/BioNTech vaccine (n = 50). Spike binding and neutralization against all three VOC was substantially reduced in the majority of samples, with the largest 4-7-fold reduction in neutralization being observed against B.1.351. While hospitalized COVID-19 patients and vaccinees maintained sufficient neutralizing titers against all three VOC, 39% of non-hospitalized patients did not neutralize B.1.351. Moreover, monoclonal neutralizing antibodies (NAbs) show sharp reductions in their binding kinetics and neutralizing potential to B.1.351 and P.1, but not to B.1.1.7. These data have implications for the degree to which pre-existing immunity can protect against subsequent infection with VOC and informs policy makers of susceptibility to globally circulating SARS-CoV-2 VOC.
Collapse
|
349
|
Zhao M, Su PY, Castro DA, Tripler TN, Hu Y, Cook M, Ko AI, Farhadian SF, Israelow B, Dela Cruz CS, Xiong Y, Sutton RE, the Yale IMPACT Research Team. Rapid, reliable, and reproducible cell fusion assay to quantify SARS-Cov-2 spike interaction with hACE2. PLoS Pathog 2021; 17:e1009683. [PMID: 34166473 PMCID: PMC8263067 DOI: 10.1371/journal.ppat.1009683] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/07/2021] [Accepted: 06/02/2021] [Indexed: 12/13/2022] Open
Abstract
COVID-19 is a global crisis of unimagined dimensions. Currently, Remedesivir is only fully licensed FDA therapeutic. A major target of the vaccine effort is the SARS-CoV-2 spike-hACE2 interaction, and assessment of efficacy relies on time consuming neutralization assay. Here, we developed a cell fusion assay based upon spike-hACE2 interaction. The system was tested by transient co-transfection of 293T cells, which demonstrated good correlation with standard spike pseudotyping for inhibition by sera and biologics. Then established stable cell lines were very well behaved and gave even better correlation with pseudotyping results, after a short, overnight co-incubation. Results with the stable cell fusion assay also correlated well with those of a live virus assay. In summary we have established a rapid, reliable, and reproducible cell fusion assay that will serve to complement the other neutralization assays currently in use, is easy to implement in most laboratories, and may serve as the basis for high throughput screens to identify inhibitors of SARS-CoV-2 virus-cell binding and entry.
Collapse
Affiliation(s)
- Min Zhao
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Pei-Yi Su
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Danielle A. Castro
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Therese N. Tripler
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Yingxia Hu
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Matthew Cook
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Albert I. Ko
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Shelli F. Farhadian
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Benjamin Israelow
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Charles S. Dela Cruz
- Department of Internal Medicine, Section of Pulmonary and Critical Care Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Yong Xiong
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Richard E. Sutton
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, United States of America
| | | |
Collapse
|
350
|
Yamin R, Jones AT, Hoffmann HH, Kao KS, Francis RL, Sheahan TP, Baric RS, Rice CM, Ravetch JV, Bournazos S. Fc-engineered antibody therapeutics with improved efficacy against COVID-19. RESEARCH SQUARE 2021:rs.3.rs-555612. [PMID: 34075373 PMCID: PMC8168397 DOI: 10.21203/rs.3.rs-555612/v1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Monoclonal antibodies (mAbs) with neutralizing activity against SARS-CoV-2 have demonstrated clinical benefit in cases of mild to moderate SARS-CoV-2 infection, substantially reducing the risk for hospitalization and severe disease1-4. Treatment generally requires the administration of high doses of these mAbs with limited efficacy in preventing disease complications or mortality among hospitalized COVID-19 patients5. Here we report the development and evaluation of Fc-optimized anti-SARS-CoV-2 mAbs with superior potency to prevent or treat COVID-19 disease. In several animal models of COVID-19 disease6,7, we demonstrate that selective engagement of activating FcγRs results in improved efficacy in both preventing and treating disease-induced weight loss and mortality, significantly reducing the dose required to confer full protection upon SARS-CoV-2 challenge and treatment of pre-infected animals. Our results highlight the importance of FcγR pathways in driving antibody-mediated antiviral immunity, while excluding any pathogenic or disease-enhancing effects of FcγR engagement of anti-SARS-CoV-2 antibodies upon infection. These findings have important implications for the development of Fc-engineered mAbs with optimal Fc effector function and improved clinical efficacy against COVID-19 disease.
Collapse
Affiliation(s)
- Rachel Yamin
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY
| | - Andrew T Jones
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY
| | | | - Kevin S Kao
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY
| | - Rebecca L Francis
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY
| | - Timothy P Sheahan
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY
| | - Stylianos Bournazos
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY
| |
Collapse
|