1
|
Sartingen N, Stürmer V, Kaltenböck M, Müller TG, Schnitzler P, Kreshuk A, Kräusslich HG, Merle U, Mücksch F, Müller B, Pape C, Laketa V. Multiplex Microscopy Assay for Assessment of Therapeutic and Serum Antibodies against Emerging Pathogens. Viruses 2024; 16:1473. [PMID: 39339949 PMCID: PMC11437451 DOI: 10.3390/v16091473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
The emergence of novel pathogens, exemplified recently by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), highlights the need for rapidly deployable and adaptable diagnostic assays to assess their impact on human health and guide public health responses in future pandemics. In this study, we developed an automated multiplex microscopy assay coupled with machine learning-based analysis for antibody detection. To achieve multiplexing and simultaneous detection of multiple viral antigens, we devised a barcoding strategy utilizing a panel of HeLa-based cell lines. Each cell line expressed a distinct viral antigen, along with a fluorescent protein exhibiting a unique subcellular localization pattern for cell classification. Our robust, cell segmentation and classification algorithm, combined with automated image acquisition, ensured compatibility with a high-throughput approach. As a proof of concept, we successfully applied this approach for quantitation of immunoreactivity against different variants of SARS-CoV-2 spike and nucleocapsid proteins in sera of patients or vaccinees, as well as for the study of selective reactivity of monoclonal antibodies. Importantly, our system can be rapidly adapted to accommodate other SARS-CoV-2 variants as well as any antigen of a newly emerging pathogen, thereby representing an important resource in the context of pandemic preparedness.
Collapse
Affiliation(s)
- Nuno Sartingen
- Department of Infectious Diseases, Virology, Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany; (N.S.); (V.S.); (M.K.); (T.G.M.); (P.S.); (H.-G.K.); (F.M.); (B.M.)
| | - Vanessa Stürmer
- Department of Infectious Diseases, Virology, Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany; (N.S.); (V.S.); (M.K.); (T.G.M.); (P.S.); (H.-G.K.); (F.M.); (B.M.)
| | - Matthias Kaltenböck
- Department of Infectious Diseases, Virology, Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany; (N.S.); (V.S.); (M.K.); (T.G.M.); (P.S.); (H.-G.K.); (F.M.); (B.M.)
| | - Thorsten G. Müller
- Department of Infectious Diseases, Virology, Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany; (N.S.); (V.S.); (M.K.); (T.G.M.); (P.S.); (H.-G.K.); (F.M.); (B.M.)
| | - Paul Schnitzler
- Department of Infectious Diseases, Virology, Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany; (N.S.); (V.S.); (M.K.); (T.G.M.); (P.S.); (H.-G.K.); (F.M.); (B.M.)
| | - Anna Kreshuk
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany;
| | - Hans-Georg Kräusslich
- Department of Infectious Diseases, Virology, Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany; (N.S.); (V.S.); (M.K.); (T.G.M.); (P.S.); (H.-G.K.); (F.M.); (B.M.)
- German Center for Infection Research (DZIF), Partner Site Heidelberg, 69120 Heidelberg, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Frauke Mücksch
- Department of Infectious Diseases, Virology, Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany; (N.S.); (V.S.); (M.K.); (T.G.M.); (P.S.); (H.-G.K.); (F.M.); (B.M.)
| | - Barbara Müller
- Department of Infectious Diseases, Virology, Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany; (N.S.); (V.S.); (M.K.); (T.G.M.); (P.S.); (H.-G.K.); (F.M.); (B.M.)
| | - Constantin Pape
- Institute of Computer Science, Göttingen University, 37073 Göttingen, Germany;
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), Göttingen University, 37073 Göttingen, Germany
| | - Vibor Laketa
- Department of Infectious Diseases, Virology, Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany; (N.S.); (V.S.); (M.K.); (T.G.M.); (P.S.); (H.-G.K.); (F.M.); (B.M.)
- German Center for Infection Research (DZIF), Partner Site Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
2
|
St. Germain R, Bossard EL, Corey L, Sholukh AM. Serum concentration of antigen-specific IgG can substantially bias interpretation of antibody-dependent phagocytosis assay readout. iScience 2023; 26:107527. [PMID: 37664583 PMCID: PMC10469534 DOI: 10.1016/j.isci.2023.107527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 06/21/2023] [Accepted: 07/28/2023] [Indexed: 09/05/2023] Open
Abstract
Because virus neutralization cannot solely explain vaccine-induced, antibody-mediated protection, antibody effector functions are being considered as a potential correlate of protection (CoP). However, measuring effector functions at a fixed serum dilution for high throughput purposes makes it difficult to distinguish between the effect of serum antibody concentration and antibody properties such as epitopes, subclass, and glycosylation. To address this issue, we evaluated antibody-dependent cellular phagocytosis (ADCP) assay against SARS-CoV-2 spike. Adjustment of serum samples to the same concentration of antigen-specific IgG prior to the ADCP assay revealed concentration-independent differences in ADCP after mRNA vaccination in subjects with and without prior SARS-CoV-2 infection not detectable in assay performed with fixed serum dilution. Phagocytosis measured at different concentrations of spike-specific IgG strongly correlated with the area under the curve (AUC) indicating that ADCP assay can be performed at a standardized antibody concentration for the high throughput necessary for vaccine trial analyses.
Collapse
Affiliation(s)
- Russell St. Germain
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA
| | - Emily L. Bossard
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA
| | - Lawrence Corey
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Anton M. Sholukh
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA
| |
Collapse
|
3
|
Motsoeneng BM, Manamela NP, Kaldine H, Kgagudi P, Hermanus T, Ayres F, Makhado Z, Moyo-Gwete T, van der Mescht MA, Abdullah F, Boswell MT, Ueckermann V, Rossouw TM, Madhi SA, Moore PL, Richardson SI. Despite delayed kinetics, people living with HIV achieve equivalent antibody function after SARS-CoV-2 infection or vaccination. Front Immunol 2023; 14:1231276. [PMID: 37600825 PMCID: PMC10435738 DOI: 10.3389/fimmu.2023.1231276] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
The kinetics of Fc-mediated functions following SARS-CoV-2 infection or vaccination in people living with HIV (PLWH) are not known. We compared SARS-CoV-2 spike-specific Fc functions, binding, and neutralization in PLWH and people without HIV (PWOH) during acute infection (without prior vaccination) with either the D614G or Beta variants of SARS-CoV-2, or vaccination with ChAdOx1 nCoV-19. Antiretroviral treatment (ART)-naïve PLWH had significantly lower levels of IgG binding, neutralization, and antibody-dependent cellular phagocytosis (ADCP) compared with PLWH on ART. The magnitude of antibody-dependent cellular cytotoxicity (ADCC), complement deposition (ADCD), and cellular trogocytosis (ADCT) was differentially triggered by D614G and Beta. The kinetics of spike IgG-binding antibodies, ADCC, and ADCD were similar, irrespective of the infecting variant between PWOH and PLWH overall. However, compared with PWOH, PLWH infected with D614G had delayed neutralization and ADCP. Furthermore, Beta infection resulted in delayed ADCT, regardless of HIV status. Despite these delays, we observed improved coordination between binding and neutralizing responses and Fc functions in PLWH. In contrast to D614G infection, binding responses in PLWH following ChAdOx-1 nCoV-19 vaccination were delayed, while neutralization and ADCP had similar timing of onset, but lower magnitude, and ADCC was significantly higher than in PWOH. Overall, despite delayed and differential kinetics, PLWH on ART develop comparable responses to PWOH, supporting the prioritization of ART rollout and SARS-CoV-2 vaccination in PLWH.
Collapse
Affiliation(s)
- Boitumelo M. Motsoeneng
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Nelia P. Manamela
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Haajira Kaldine
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Prudence Kgagudi
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Tandile Hermanus
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Frances Ayres
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Zanele Makhado
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Thandeka Moyo-Gwete
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Mieke A. van der Mescht
- Department of Immunology, Faculty of Health Science, University of Pretoria, Pretoria, South Africa
| | - Fareed Abdullah
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital and University of Pretoria, Pretoria, South Africa
- South African Medical Research Council Office of AIDS and TB Research, Pretoria, South Africa
| | - Michael T. Boswell
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital and University of Pretoria, Pretoria, South Africa
| | - Veronica Ueckermann
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital and University of Pretoria, Pretoria, South Africa
| | - Theresa M. Rossouw
- Department of Immunology, Faculty of Health Science, University of Pretoria, Pretoria, South Africa
| | - Shabir A. Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Infectious Diseases and Oncology Research Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Penny L. Moore
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu Natal, Durban, South Africa
| | - Simone I. Richardson
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| |
Collapse
|
4
|
Tong X, McNamara RP, Avendaño MJ, Serrano EF, García-Salum T, Pardo-Roa C, Bertera HL, Chicz TM, Levican J, Poblete E, Salinas E, Muñoz A, Riquelme A, Alter G, Medina RA. Waning and boosting of antibody Fc-effector functions upon SARS-CoV-2 vaccination. Nat Commun 2023; 14:4174. [PMID: 37443074 PMCID: PMC10345146 DOI: 10.1038/s41467-023-39189-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 06/01/2023] [Indexed: 07/15/2023] Open
Abstract
Since the emergence of SARS-CoV-2, vaccines targeting COVID-19 have been developed with unprecedented speed and efficiency. CoronaVac, utilising an inactivated form of the COVID-19 virus and the mRNA26 based Pfizer/BNT162b2 vaccines are widely distributed. Beyond the ability of vaccines to induce production of neutralizing antibodies, they might lead to the generation of antibodies attenuating the disease by recruiting cytotoxic and opsonophagocytic functions. However, the Fc-effector functions of vaccine induced antibodies are much less studied than virus neutralization. Here, using systems serology, we follow the longitudinal Fc-effector profiles induced by CoronaVac and BNT162b2 up until five months following the two-dose vaccine regimen. Compared to BNT162b2, CoronaVac responses wane more slowly, albeit the levels remain lower than that of BNT162b2 recipients throughout the entire observation period. However, mRNA vaccine boosting of CoronaVac responses, including response to the Omicron variant, induce significantly higher peak of antibody functional responses with increased humoral breadth. In summary, we show that vaccine platform-induced humoral responses are not limited to virus neutralization but rather utilise antibody dependent effector functions. We demonstrate that this functionality wanes with different kinetics and can be rescued and expanded via boosting with subsequent homologous and heterologous vaccination.
Collapse
Affiliation(s)
- X Tong
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - R P McNamara
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - M J Avendaño
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - E F Serrano
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - T García-Salum
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) - COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - C Pardo-Roa
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) - COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - H L Bertera
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - T M Chicz
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - J Levican
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - E Poblete
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - E Salinas
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) - COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - A Muñoz
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - A Riquelme
- Advanced Interdisciplinary Rehabilitation Register (AIRR) - COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, 8331150, Chile
| | - G Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA.
| | - R A Medina
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile.
- Advanced Interdisciplinary Rehabilitation Register (AIRR) - COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile.
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, 30322, USA.
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
5
|
Sunagar R, Singh A, Kumar S. SARS-CoV-2: Immunity, Challenges with Current Vaccines, and a Novel Perspective on Mucosal Vaccines. Vaccines (Basel) 2023; 11:vaccines11040849. [PMID: 37112761 PMCID: PMC10143972 DOI: 10.3390/vaccines11040849] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
The global rollout of COVID-19 vaccines has played a critical role in reducing pandemic spread, disease severity, hospitalizations, and deaths. However, the first-generation vaccines failed to block severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and transmission, partially due to the limited induction of mucosal immunity, leading to the continuous emergence of variants of concern (VOC) and breakthrough infections. To meet the challenges from VOC, limited durability, and lack of mucosal immune response of first-generation vaccines, novel approaches are being investigated. Herein, we have discussed the current knowledge pertaining to natural and vaccine-induced immunity, and the role of the mucosal immune response in controlling SARS-CoV2 infection. We have also presented the current status of the novel approaches aimed at eliciting both mucosal and systemic immunity. Finally, we have presented a novel adjuvant-free approach to elicit effective mucosal immunity against SARS-CoV-2, which lacks the safety concerns associated with live-attenuated vaccine platforms.
Collapse
Affiliation(s)
| | - Amit Singh
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| | - Sudeep Kumar
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
6
|
Mackin SR, Desai P, Whitener BM, Karl CE, Liu M, Baric RS, Edwards DK, Chicz TM, McNamara RP, Alter G, Diamond MS. Fcγ receptor-dependent antibody effector functions are required for vaccine protection against infection by antigenic variants of SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.11.27.518117. [PMID: 36482975 PMCID: PMC9727771 DOI: 10.1101/2022.11.27.518117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Emerging SARS-CoV-2 variants with antigenic changes in the spike protein are neutralized less efficiently by serum antibodies elicited by legacy vaccines against the ancestral Wuhan-1 virus. Nonetheless, these vaccines, including mRNA-1273 and BNT162b2, retained their ability to protect against severe disease and death, suggesting that other aspects of immunity control infection in the lung. Although vaccine-elicited antibodies can bind Fc gamma receptors (FcγRs) and mediate effector functions against SARS-CoV-2 variants, and this property correlates with improved clinical COVID-19 outcome, a causal relationship between Fc effector functions and vaccine-mediated protection against infection has not been established. Here, using passive and active immunization approaches in wild-type and Fc-gamma receptor (FcγR) KO mice, we determined the requirement for Fc effector functions to protect against SARS-CoV-2 infection. The antiviral activity of passively transferred immune serum was lost against multiple SARS-CoV-2 strains in mice lacking expression of activating FcγRs, especially murine FcγR III (CD16), or depleted of alveolar macrophages. After immunization with the preclinical mRNA-1273 vaccine, protection against Omicron BA.5 infection in the respiratory tract also was lost in mice lacking FcγR III. Our passive and active immunization studies in mice suggest that Fc-FcγR engagement and alveolar macrophages are required for vaccine-induced antibody-mediated protection against infection by antigenically changed SARS-CoV-2 variants, including Omicron strains.
Collapse
Affiliation(s)
- Samantha R. Mackin
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO
| | - Pritesh Desai
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Bradley M. Whitener
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Courtney E. Karl
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| | - Meizi Liu
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC
| | | | | | | | - Galit Alter
- Moderna, Inc., Cambridge MA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA
- Andrew M. and Jane M. Bursky the Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
- Andrew M. and Jane M. Bursky the Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
7
|
An Fc variant with two mutations confers prolonged serum half-life and enhanced effector functions on IgG antibodies. Exp Mol Med 2022; 54:1850-1861. [PMID: 36319752 PMCID: PMC9628495 DOI: 10.1038/s12276-022-00870-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/07/2022] Open
Abstract
The pH-selective interaction between the immunoglobulin G (IgG) fragment crystallizable region (Fc region) and the neonatal Fc receptor (FcRn) is critical for prolonging the circulating half-lives of IgG molecules through intracellular trafficking and recycling. By using directed evolution, we successfully identified Fc mutations that improve the pH-dependent binding of human FcRn and prolong the serum persistence of a model IgG antibody and an Fc-fusion protein. Strikingly, trastuzumab-PFc29 and aflibercept-PFc29, a model therapeutic IgG antibody and an Fc-fusion protein, respectively, when combined with our engineered Fc (Q311R/M428L), both exhibited significantly higher serum half-lives in human FcRn transgenic mice than their counterparts with wild-type Fc. Moreover, in a cynomolgus monkey model, trastuzumab-PFc29 displayed a superior pharmacokinetic profile to that of both trastuzumab-YTE and trastuzumab-LS, which contain the well-validated serum half-life extension Fcs YTE (M252Y/S254T/T256E) and LS (M428L/N434S), respectively. Furthermore, the introduction of two identified mutations of PFc29 (Q311R/M428L) into the model antibodies enhanced both complement-dependent cytotoxicity and antibody-dependent cell-mediated cytotoxicity activity, which are triggered by the association between IgG Fc and Fc binding ligands and are critical for clearing cancer cells. In addition, the effector functions could be turned off by combining the two mutations of PFc29 with effector function-silencing mutations, but the antibodies maintained their excellent pH-dependent human FcRn binding profile. We expect our Fc variants to be an excellent tool for enhancing the pharmacokinetic profiles and potencies of various therapeutic antibodies and Fc-fusion proteins.
Collapse
|
8
|
Xu S, Carpenter MC, Spreng RL, Neidich SD, Sarkar S, Tenney D, Goodman D, Sawant S, Jha S, Dunn B, Juliana McElrath M, Bekker V, Mudrak SV, Flinko R, Lewis GK, Ferrari G, Tomaras GD, Shen X, Ackerman ME. Impact of adjuvants on the biophysical and functional characteristics of HIV vaccine-elicited antibodies in humans. NPJ Vaccines 2022; 7:90. [PMID: 35927399 PMCID: PMC9352797 DOI: 10.1038/s41541-022-00514-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/01/2022] [Indexed: 01/14/2023] Open
Abstract
Adjuvants can alter the magnitude, characteristics, and persistence of the humoral response to protein vaccination. HIV vaccination might benefit from tailored adjuvant choice as raising a durable and protective response to vaccination has been exceptionally challenging. Analysis of trials of partially effective HIV vaccines have identified features of the immune response that correlate with decreased risk, including high titers of V1V2-binding IgG and IgG3 responses with low titers of V1V2-binding IgA responses and enhanced Fc effector functions, notably antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). However, there has been limited opportunity to compare the effect of different adjuvants on these activities in humans. Here, samples from the AVEG015 study, a phase 1 trial in which participants (n = 112) were immunized with gp120SF-2 and one of six different adjuvants or combinations thereof were assessed for antibody titer, biophysical features, and diverse effector functions. Three adjuvants, MF59 + MTP-PE, SAF/2, and SAF/2 + MDP, increased the peak magnitude and durability of antigen-specific IgG3, IgA, FcγR-binding responses and ADCP activity, as compared to alum. While multiple adjuvants increased the titer of IgG, IgG3, and IgA responses, none consistently altered the balance of IgG to IgA or IgG3 to IgA. Linear regression analysis identified biophysical features including gp120-specific IgG and FcγR-binding responses that could predict functional activity, and network analysis identified coordinated aspects of the humoral response. These analyses reveal the ability of adjuvants to drive the character and function of the humoral response despite limitations of small sample size and immune variability in this human clinical trial.
Collapse
Affiliation(s)
- Shiwei Xu
- Quantitative Biomedical Science Program, Dartmouth College, Hanover, NH, USA
| | | | - Rachel L Spreng
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Scott D Neidich
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Sharanya Sarkar
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - DeAnna Tenney
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Derrick Goodman
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Sheetal Sawant
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Shalini Jha
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Brooke Dunn
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Departments of Laboratory Medicine and Medicine, University of Washington, Seattle, WA, USA
| | - Valerie Bekker
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Sarah V Mudrak
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Robin Flinko
- Division of Vaccine Research, The Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - George K Lewis
- Division of Vaccine Research, The Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Guido Ferrari
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Georgia D Tomaras
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Xiaoying Shen
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA.
| | - Margaret E Ackerman
- Quantitative Biomedical Science Program, Dartmouth College, Hanover, NH, USA.
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
9
|
Claireaux M, Caniels TG, de Gast M, Han J, Guerra D, Kerster G, van Schaik BDC, Jongejan A, Schriek AI, Grobben M, Brouwer PJM, van der Straten K, Aldon Y, Capella-Pujol J, Snitselaar JL, Olijhoek W, Aartse A, Brinkkemper M, Bontjer I, Burger JA, Poniman M, Bijl TPL, Torres JL, Copps J, Martin IC, de Taeye SW, de Bree GJ, Ward AB, Sliepen K, van Kampen AHC, Moerland PD, Sanders RW, van Gils MJ. A public antibody class recognizes an S2 epitope exposed on open conformations of SARS-CoV-2 spike. Nat Commun 2022; 13:4539. [PMID: 35927266 PMCID: PMC9352689 DOI: 10.1038/s41467-022-32232-0] [Citation(s) in RCA: 40] [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: 11/30/2021] [Accepted: 07/22/2022] [Indexed: 12/21/2022] Open
Abstract
Delineating the origins and properties of antibodies elicited by SARS-CoV-2 infection and vaccination is critical for understanding their benefits and potential shortcomings. Therefore, we investigate the SARS-CoV-2 spike (S)-reactive B cell repertoire in unexposed individuals by flow cytometry and single-cell sequencing. We show that ∼82% of SARS-CoV-2 S-reactive B cells harbor a naive phenotype, which represents an unusually high fraction of total human naive B cells (∼0.1%). Approximately 10% of these naive S-reactive B cells share an IGHV1-69/IGKV3-11 B cell receptor pairing, an enrichment of 18-fold compared to the complete naive repertoire. Following SARS-CoV-2 infection, we report an average 37-fold enrichment of IGHV1-69/IGKV3-11 B cell receptor pairing in the S-reactive memory B cells compared to the unselected memory repertoire. This class of B cells targets a previously undefined non-neutralizing epitope on the S2 subunit that becomes exposed on S proteins used in approved vaccines when they transition away from the native pre-fusion state because of instability. These findings can help guide the improvement of SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Mathieu Claireaux
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Tom G Caniels
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Marlon de Gast
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Julianna Han
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Denise Guerra
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Gius Kerster
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Barbera D C van Schaik
- Bioinformatics Laboratory, Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam Institute for Public Health, Amsterdam, the Netherlands
| | - Aldo Jongejan
- Bioinformatics Laboratory, Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam Institute for Public Health, Amsterdam, the Netherlands
| | - Angela I Schriek
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Marloes Grobben
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Philip J M Brouwer
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Karlijn van der Straten
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Yoann Aldon
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Joan Capella-Pujol
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Jonne L Snitselaar
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Wouter Olijhoek
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Aafke Aartse
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Mitch Brinkkemper
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Ilja Bontjer
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Judith A Burger
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Meliawati Poniman
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Tom P L Bijl
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Jonathan L Torres
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jeffrey Copps
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Isabel Cuella Martin
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Steven W de Taeye
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Godelieve J de Bree
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Kwinten Sliepen
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Antoine H C van Kampen
- Bioinformatics Laboratory, Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam Institute for Public Health, Amsterdam, the Netherlands
| | - Perry D Moerland
- Bioinformatics Laboratory, Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam Institute for Public Health, Amsterdam, the Netherlands
| | - Rogier W Sanders
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands.
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands.
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA.
| | - Marit J van Gils
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam, the Netherlands.
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands.
| |
Collapse
|
10
|
Nikitin PA, DiMuzio JM, Dowling JP, Patel NB, Bingaman-Steele JL, Heimbach BC, Henriquez N, Nicolescu C, Polley A, Sikorski EL, Howanski RJ, Nath M, Shukla H, Scheaffer SM, Finn JP, Liang LF, Smith T, Storm N, McKay LGA, Johnson RI, Malsick LE, Honko AN, Griffiths A, Diamond MS, Sarma P, Geising DH, Morin MJ, Robinson MK. IMM-BCP-01, a patient-derived anti-SARS-CoV-2 antibody cocktail, is active across variants of concern including Omicron BA.1 and BA.2. Sci Immunol 2022; 7:eabl9943. [PMID: 35771946 PMCID: PMC9273042 DOI: 10.1126/sciimmunol.abl9943] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Monoclonal antibodies are an efficacious therapy against SARS-CoV-2. However, rapid viral mutagenesis, led to escape from most of these therapies, outlining the need for an antibody cocktail with a broad neutralizing potency. Using an unbiased interrogation of the memory B cell repertoire of convalescent COVID-19 patients, we identified human antibodies with broad antiviral activity in vitro and efficacy in vivo against all tested SARS-CoV-2 variants of concern, including Delta, Omicron BA.1 and BA.2. Here, we describe an antibody cocktail IMM-BCP-01, that consists of three patient-derived broadly neutralizing antibodies directed at non-overlapping surfaces on the SARS-CoV-2 spike protein. Two antibodies, IMM20184 and IMM20190, directly blocked Spike binding to the ACE2 receptor. Binding of the third antibody, IMM20253, to its cryptic epitope on the outer surface of RBD, altered the conformation of the Spike Trimer, promoting release of Spike monomers. These antibodies decreased Omicron SARS-CoV-2 infection in the lungs of Syrian golden hamsters in vivo, and potently induced antiviral effector response in vitro, including phagocytosis, ADCC, and complement pathway activation. Our pre-clinical data demonstrated that the three antibody cocktail IMM-BCP-01 could be a promising means for preventing or treating infection of SARS-CoV-2 variants of concern, including Omicron BA.1 and BA.2, in susceptible individuals.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Suzanne M Scheaffer
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | - Nadia Storm
- Department of Microbiology, Boston University School of Medicine and National Emerging Infectious Diseases Laboratories, Boston, MA, USA
| | - Lindsay G A McKay
- Department of Microbiology, Boston University School of Medicine and National Emerging Infectious Diseases Laboratories, Boston, MA, USA
| | - Rebecca I Johnson
- Department of Microbiology, Boston University School of Medicine and National Emerging Infectious Diseases Laboratories, Boston, MA, USA
| | - Lauren E Malsick
- Department of Microbiology, Boston University School of Medicine and National Emerging Infectious Diseases Laboratories, Boston, MA, USA
| | - Anna N Honko
- Department of Microbiology, Boston University School of Medicine and National Emerging Infectious Diseases Laboratories, Boston, MA, USA
| | - Anthony Griffiths
- Department of Microbiology, Boston University School of Medicine and National Emerging Infectious Diseases Laboratories, Boston, MA, USA
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
11
|
Sefik E, Qu R, Junqueira C, Kaffe E, Mirza H, Zhao J, Brewer JR, Han A, Steach HR, Israelow B, Blackburn HN, Velazquez SE, Chen YG, Halene S, Iwasaki A, Meffre E, Nussenzweig M, Lieberman J, Wilen CB, Kluger Y, Flavell RA. Inflammasome activation in infected macrophages drives COVID-19 pathology. Nature 2022; 606:585-593. [PMID: 35483404 PMCID: PMC9288243 DOI: 10.1038/s41586-022-04802-1] [Citation(s) in RCA: 326] [Impact Index Per Article: 108.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 04/25/2022] [Indexed: 01/18/2023]
Abstract
Severe COVID-19 is characterized by persistent lung inflammation, inflammatory cytokine production, viral RNA and a sustained interferon (IFN) response, all of which are recapitulated and required for pathology in the SARS-CoV-2-infected MISTRG6-hACE2 humanized mouse model of COVID-19, which has a human immune system1-20. Blocking either viral replication with remdesivir21-23 or the downstream IFN-stimulated cascade with anti-IFNAR2 antibodies in vivo in the chronic stages of disease attenuates the overactive immune inflammatory response, especially inflammatory macrophages. Here we show that SARS-CoV-2 infection and replication in lung-resident human macrophages is a critical driver of disease. In response to infection mediated by CD16 and ACE2 receptors, human macrophages activate inflammasomes, release interleukin 1 (IL-1) and IL-18, and undergo pyroptosis, thereby contributing to the hyperinflammatory state of the lungs. Inflammasome activation and the accompanying inflammatory response are necessary for lung inflammation, as inhibition of the NLRP3 inflammasome pathway reverses chronic lung pathology. Notably, this blockade of inflammasome activation leads to the release of infectious virus by the infected macrophages. Thus, inflammasomes oppose host infection by SARS-CoV-2 through the production of inflammatory cytokines and suicide by pyroptosis to prevent a productive viral cycle.
Collapse
Affiliation(s)
- Esen Sefik
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Rihao Qu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Computational Biology and Bioinformatics Program, Yale University, New Haven, CT, USA
| | - Caroline Junqueira
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Eleanna Kaffe
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Haris Mirza
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Jun Zhao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - J Richard Brewer
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ailin Han
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Holly R Steach
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Benjamin Israelow
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Holly N Blackburn
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Sofia E Velazquez
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Y Grace Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie Halene
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Section of Hematology, Yale Cancer Center and Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Eric Meffre
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Michel Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Craig B Wilen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Yuval Kluger
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Program of Applied Mathematics, Yale University, New Haven, CT, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
12
|
Sefik E, Israelow B, Mirza H, Zhao J, Qu R, Kaffe E, Song E, Halene S, Meffre E, Kluger Y, Nussenzweig M, Wilen CB, Iwasaki A, Flavell RA. A humanized mouse model of chronic COVID-19. Nat Biotechnol 2022; 40:906-920. [PMID: 34921308 PMCID: PMC9203605 DOI: 10.1038/s41587-021-01155-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 11/05/2021] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease that can present as an uncontrolled, hyperactive immune response, causing severe immunological injury. Existing rodent models do not recapitulate the sustained immunopathology of patients with severe disease. Here we describe a humanized mouse model of COVID-19 that uses adeno-associated virus to deliver human ACE2 to the lungs of humanized MISTRG6 mice. This model recapitulates innate and adaptive human immune responses to severe acute respiratory syndrome coronavirus 2 infection up to 28 days after infection, with key features of chronic COVID-19, including weight loss, persistent viral RNA, lung pathology with fibrosis, a human inflammatory macrophage response, a persistent interferon-stimulated gene signature and T cell lymphopenia. We used this model to study two therapeutics on immunopathology, patient-derived antibodies and steroids and found that the same inflammatory macrophages crucial to containing early infection later drove immunopathology. This model will enable evaluation of COVID-19 disease mechanisms and treatments.
Collapse
Affiliation(s)
- Esen Sefik
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Benjamin Israelow
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Haris Mirza
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Jun Zhao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Rihao Qu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Eleanna Kaffe
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Eric Song
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie Halene
- Section of Hematology, Yale Cancer Center and Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Eric Meffre
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Yuval Kluger
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Michel Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Craig B Wilen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
13
|
Effective Interferon Lambda Treatment Regimen To Control Lethal MERS-CoV Infection in Mice. J Virol 2022; 96:e0036422. [PMID: 35588276 DOI: 10.1128/jvi.00364-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Effective broad-spectrum antivirals are critical to prevent and control emerging human coronavirus (hCoV) infections. Despite considerable progress made toward identifying and evaluating several synthetic broad-spectrum antivirals against hCoV infections, a narrow therapeutic window has limited their success. Enhancing the endogenous interferon (IFN) and IFN-stimulated gene (ISG) response is another antiviral strategy that has been known for decades. However, the side effects of pegylated type-I IFNs (IFN-Is) and the proinflammatory response detected after delayed IFN-I therapy have discouraged their clinical use. In contrast to IFN-Is, IFN-λ, a dominant IFN at the epithelial surface, has been shown to be less proinflammatory. Consequently, we evaluated the prophylactic and therapeutic efficacy of IFN-λ in hCoV-infected airway epithelial cells and mice. Human primary airway epithelial cells treated with a single dose of IFN-I (IFN-α) and IFN-λ showed similar ISG expression, whereas cells treated with two doses of IFN-λ expressed elevated levels of ISG compared to that of IFN-α-treated cells. Similarly, mice treated with two doses of IFN-λ were better protected than mice that received a single dose, and a combination of prophylactic and delayed therapeutic regimens completely protected mice from a lethal Middle East respiratory syndrome CoV (MERS-CoV) infection. A two-dose IFN-λ regimen significantly reduced lung viral titers and inflammatory cytokine levels with marked improvement in lung inflammation. Collectively, we identified an effective regimen for IFN-λ use and demonstrated the protective efficacy of IFN-λ in MERS-CoV-infected mice. IMPORTANCE Effective antiviral agents are urgently required to prevent and treat individuals infected with SARS-CoV-2 and other emerging viral infections. The COVID-19 pandemic has catapulted our efforts to identify, develop, and evaluate several antiviral agents. However, a narrow therapeutic window has limited the protective efficacy of several broad-spectrum and CoV-specific antivirals. IFN-λ is an antiviral agent of interest due to its ability to induce a robust endogenous antiviral state and low levels of inflammation. Here, we evaluated the protective efficacy and effective treatment regimen of IFN-λ in mice infected with a lethal dose of MERS-CoV. We show that while prophylactic and early therapeutic IFN-λ administration is protective, delayed treatment is detrimental. Notably, a combination of prophylactic and delayed therapeutic administration of IFN-λ protected mice from severe MERS. Our results highlight the prophylactic and therapeutic use of IFN-λ against lethal hCoV and likely other viral lung infections.
Collapse
|
14
|
van der Straten K, van Gils MJ, de Taeye SW, de Bree GJ. Optimization of Anti-SARS-CoV-2 Neutralizing Antibody Therapies: Roadmap to Improve Clinical Effectiveness and Implementation. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:867982. [PMID: 35419561 PMCID: PMC8996231 DOI: 10.3389/fmedt.2022.867982] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/07/2022] [Indexed: 12/11/2022] Open
Abstract
One of the major breakthroughs to combat the current Coronavirus Disease 2019 (COVID-19) pandemic has been the development of highly effective vaccines against the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Still, alternatives are needed for individuals who are at high risk of developing severe COVID-19 and are not protected by vaccination. Monoclonal antibodies against the spike protein of SARS-CoV-2 have been shown to be effective as prophylaxis and treatment against COVID-19. However, the emergence of variants of concern (VOCs) challenges the efficacy of antibody therapies. This review describes the neutralization resistance of the clinically-approved monoclonal antibody therapies against the Alpha (B.1.1.7), Beta (B.1.351), Gamma (P1), Delta (B.1.617.2), and the Omicron (B.1.1.529) variants. To guide the development of monoclonal antibody therapies and to anticipate on the continuous evolution of SARS-CoV-2, we highlight different strategies to broaden the antibody activity by targeting more conserved epitopes and/or simultaneously targeting multiple sites of vulnerability of the virus. This review further describes the contribution of antibody Fc effector functions to optimize the antibody efficacy. In addition, the main route of SARS-CoV-2 antibody administration is currently intravenously and dictates a monthly injection when used as prophylactic. Therefore, we discusses the concept of long-acting antibodies (LAABs) and non-intravenously routes of antibody administration in order to broaden the clinical applicability of antibody therapies.
Collapse
Affiliation(s)
- Karlijn van der Straten
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Internal Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Marit J. van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Steven W. de Taeye
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Godelieve J. de Bree
- Department of Internal Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
15
|
Inchingolo AD, Dipalma G, Inchingolo AM, Malcangi G, Santacroce L, D’Oria MT, Isacco CG, Bordea IR, Candrea S, Scarano A, Morandi B, Del Fabbro M, Farronato M, Tartaglia GM, Balzanelli MG, Ballini A, Nucci L, Lorusso F, Taschieri S, Inchingolo F. The 15-Months Clinical Experience of SARS-CoV-2: A Literature Review of Therapies and Adjuvants. Antioxidants (Basel) 2021; 10:881. [PMID: 34072708 PMCID: PMC8226610 DOI: 10.3390/antiox10060881] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus responsible for the coronavirus disease of 2019 (COVID-19) that emerged in December 2019 in Wuhan, China, and rapidly spread worldwide, with a daily increase in confirmed cases and infection-related deaths. The World Health Organization declared a pandemic on the 11th of March 2020. COVID-19 presents flu-like symptoms that become severe in high-risk medically compromised subjects. The aim of this study was to perform an updated overview of the treatments and adjuvant protocols for COVID-19. METHODS A systematic literature search of databases was performed (MEDLINE PubMed, Google Scholar, UpToDate, Embase, and Web of Science) using the keywords: "COVID-19", "2019-nCoV", "coronavirus" and "SARS-CoV-2" (date range: 1 January 2019 to 31st October 2020), focused on clinical features and treatments. RESULTS The main treatments retrieved were antivirals, antimalarials, convalescent plasma, immunomodulators, corticosteroids, anticoagulants, and mesenchymal stem cells. Most of the described treatments may provide benefits to COVID-19 subjects, but no one protocol has definitively proven its efficacy. CONCLUSIONS While many efforts are being spent worldwide in research aimed at identifying early diagnostic methods and evidence-based effective treatments, mass vaccination is thought to be the best option against this disease in the near future.
Collapse
Affiliation(s)
- Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
| | - Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
| | - Luigi Santacroce
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
| | - Maria Teresa D’Oria
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
- Department of Medical and Biological Sciences, University of Udine, Via delle Scienze, 206, 33100 Udine, Italy
| | - Ciro Gargiulo Isacco
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
- Research at Human Stem Cells Research Center HSC, Ho Chi Minh 70000, Vietnam
- Embryology and Regenerative Medicine and Immunology, Pham Chau Trinh University of Medicine Hoi An, Hoi An 70000, Vietnam
| | - Ioana Roxana Bordea
- Department of Oral Rehabilitation, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania;
| | - Sebastian Candrea
- Department of Oral Rehabilitation, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania;
- Department of Pedodontics, County Hospital Cluj-Napoca, 400000 Cluj-Napoca, Romania
| | - Antonio Scarano
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Benedetta Morandi
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy; (B.M.); (M.D.F.); (M.F.); (G.M.T.); (S.T.)
- Dental Clinic, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy
| | - Massimo Del Fabbro
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy; (B.M.); (M.D.F.); (M.F.); (G.M.T.); (S.T.)
- Dental Clinic, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy
| | - Marco Farronato
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy; (B.M.); (M.D.F.); (M.F.); (G.M.T.); (S.T.)
- UOC Maxillo-Facial Surgery and Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Gianluca Martino Tartaglia
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy; (B.M.); (M.D.F.); (M.F.); (G.M.T.); (S.T.)
- UOC Maxillo-Facial Surgery and Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Mario Giosuè Balzanelli
- SET-118, Department of Pre-Hospital and Emergency-San Giuseppe Moscati Hospital, 74100 Taranto, Italy;
| | - Andrea Ballini
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Campus Universitario, University of Bari, 70125 Bari, Italy;
- Department of Precision Medicine, University of Campania, 80138 Naples, Italy
| | - Ludovica Nucci
- Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, 80100 Naples, Italy;
| | - Felice Lorusso
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Silvio Taschieri
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy; (B.M.); (M.D.F.); (M.F.); (G.M.T.); (S.T.)
- Dental Clinic, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy
- Department of Oral Surgery, Institute of Dentistry, I. M. Sechenov First Moscow State Medical University, 119146 Moscow, Russia
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (G.D.); (A.M.I.); (L.S.); (M.T.D.); (C.G.I.); (F.I.)
| |
Collapse
|
16
|
Keeler SP, Fox JM. Requirement of Fc-Fc Gamma Receptor Interaction for Antibody-Based Protection against Emerging Virus Infections. Viruses 2021; 13:v13061037. [PMID: 34072720 PMCID: PMC8226613 DOI: 10.3390/v13061037] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/13/2022] Open
Abstract
Identification of therapeutics against emerging and re-emerging viruses remains a continued priority that is only reinforced by the recent SARS-CoV-2 pandemic. Advances in monoclonal antibody (mAb) isolation, characterization, and production make it a viable option for rapid treatment development. While mAbs are traditionally screened and selected based on potency of neutralization in vitro, it is clear that additional factors contribute to the in vivo efficacy of a mAb beyond viral neutralization. These factors include interactions with Fc receptors (FcRs) and complement that can enhance neutralization, clearance of infected cells, opsonization of virions, and modulation of the innate and adaptive immune response. In this review, we discuss recent studies, primarily using mouse models, that identified a role for Fc-FcγR interactions for optimal antibody-based protection against emerging and re-emerging virus infections.
Collapse
Affiliation(s)
- Shamus P. Keeler
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Julie M. Fox
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Correspondence:
| |
Collapse
|
17
|
Atyeo C, DeRiso EA, Davis C, Bordt EA, DeGuzman RM, Shook LL, Yonker LM, Fasano A, Akinwunmi B, Lauffenburger DA, Elovitz MA, Gray KJ, Edlow AG, Alter G. COVID-19 mRNA vaccines drive differential Fc-functional profiles in pregnant, lactating, and non-pregnant women. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.04.04.438404. [PMID: 33851165 PMCID: PMC8043455 DOI: 10.1101/2021.04.04.438404] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Significant immunological changes occur throughout pregnancy to tolerize the mother and allow growth of the fetal graft. However, additional local and systemic immunological adaptations also occur, allowing the maternal immune system to continue to protect the dyad against foreign invaders both during pregnancy and after birth through lactation. This fine balance of tolerance and immunity, along with physiological and hormonal changes, contribute to increased susceptibility to particular infections in pregnancy, including more severe COVID-19 disease. Whether these changes also make pregnant women less responsive to vaccination or induce altered immune responses to vaccination remains incompletely understood. To holistically define potential changes in vaccine response during pregnancy and lactation, we deeply profiled the humoral vaccine response in a group of pregnant and lactating women and non-pregnant age-matched controls. Vaccine-specific titers were comparable, albeit slightly lower, between pregnant and lactating women, compared to non-pregnant controls. Among pregnant women, we found higher antibody titers and functions in those vaccinated with the Moderna vaccine. FcR-binding and antibody effector functions were induced with delayed kinetics in both pregnant and lactating women compared to non-pregnant women. Antibody boosting resulted in high FcR-binding titers in breastmilk. These data point to an immune resistance to generate highly inflammatory antibodies during pregnancy and lactation, and a critical need to follow prime/boost timelines in this vulnerable population to ensure full immunity is attained.
Collapse
|
18
|
Greaney AJ, Starr TN, Barnes CO, Weisblum Y, Schmidt F, Caskey M, Gaebler C, Cho A, Agudelo M, Finkin S, Wang Z, Poston D, Muecksch F, Hatziioannou T, Bieniasz PD, Robbiani DF, Nussenzweig MC, Bjorkman PJ, Bloom JD. Mutational escape from the polyclonal antibody response to SARS-CoV-2 infection is largely shaped by a single class of antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.03.17.435863. [PMID: 33758856 PMCID: PMC7987015 DOI: 10.1101/2021.03.17.435863] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Monoclonal antibodies targeting a variety of epitopes have been isolated from individuals previously infected with SARS-CoV-2, but the relative contributions of these different antibody classes to the polyclonal response remains unclear. Here we use a yeast-display system to map all mutations to the viral spike receptor-binding domain (RBD) that escape binding by representatives of three potently neutralizing classes of anti-RBD antibodies with high-resolution structures. We compare the antibody-escape maps to similar maps for convalescent polyclonal plasma, including plasma from individuals from whom some of the antibodies were isolated. The plasma-escape maps most closely resemble those of a single class of antibodies that target an epitope on the RBD that includes site E484. Therefore, although the human immune system can produce antibodies that target diverse RBD epitopes, in practice the polyclonal response to infection is dominated by a single class of antibodies targeting an epitope that is already undergoing rapid evolution.
Collapse
Affiliation(s)
- Allison J. Greaney
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Genome Sciences & Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Tyler N. Starr
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Christopher O. Barnes
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Yiska Weisblum
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065, USA
| | - Fabian Schmidt
- 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
| | - Christian Gaebler
- 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
| | - Marianna Agudelo
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Shlomo Finkin
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Zijun Wang
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Daniel Poston
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065, USA
| | - Frauke Muecksch
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065, USA
| | | | - Paul D. Bieniasz
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065, USA
| | - Davide F. Robbiani
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
- Institute for Research in Biomedicine, Universita della Svizzera italiana (USI), 6500 Bellinzona, Switzerland
| | - Michel C. Nussenzweig
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Pamela J. Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jesse D. Bloom
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
19
|
Tauzin A, Nayrac M, Benlarbi M, Gong SY, Gasser R, Beaudoin-Bussières G, Brassard N, Laumaea A, Vézina D, Prévost J, Anand SP, Bourassa C, Gendron-Lepage G, Medjahed H, Goyette G, Niessl J, Tastet O, Gokool L, Morrisseau C, Arlotto P, Stamatatos L, McGuire AT, Larochelle C, Uchil P, Lu M, Mothes W, Serres GD, Moreira S, Roger M, Richard J, Martel-Laferrière V, Duerr R, Tremblay C, Kaufmann DE, Finzi A. A single BNT162b2 mRNA dose elicits antibodies with Fc-mediated effector functions and boost pre-existing humoral and T cell responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33758857 DOI: 10.1101/2021.03.18.435972] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The standard dosing of the Pfizer/BioNTech BNT162b2 mRNA vaccine validated in clinical trials includes two doses administered three weeks apart. While the decision by some public health authorities to space the doses because of limiting supply has raised concerns about vaccine efficacy, data indicate that a single dose is up to 90% effective starting 14 days after its administration. We analyzed humoral and T cells responses three weeks after a single dose of this mRNA vaccine. Despite the proven efficacy of the vaccine at this time point, no neutralizing activity were elicited in SARS-CoV-2 naïve individuals. However, we detected strong anti-receptor binding domain (RBD) and Spike antibodies with Fc-mediated effector functions and cellular responses dominated by the CD4 + T cell component. A single dose of this mRNA vaccine to individuals previously infected by SARS-CoV-2 boosted all humoral and T cell responses measured, with strong correlations between T helper and antibody immunity. Neutralizing responses were increased in both potency and breadth, with distinctive capacity to neutralize emerging variant strains. Our results highlight the importance of vaccinating uninfected and previously-infected individuals and shed new light into the potential role of Fc-mediated effector functions and T cell responses in vaccine efficacy. They also provide support to spacing the doses of two-vaccine regimens to vaccinate a larger pool of the population in the context of vaccine scarcity against SARS-CoV-2.
Collapse
|
20
|
Inchingolo AD, Inchingolo AM, Bordea IR, Malcangi G, Xhajanka E, Scarano A, Lorusso F, Farronato M, Tartaglia GM, Isacco CG, Marinelli G, D’Oria MT, Hazballa D, Santacroce L, Ballini A, Contaldo M, Inchingolo F, Dipalma G. SARS-CoV-2 Disease Adjuvant Therapies and Supplements Breakthrough for the Infection Prevention. Microorganisms 2021; 9:525. [PMID: 33806624 PMCID: PMC7999785 DOI: 10.3390/microorganisms9030525] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/28/2021] [Accepted: 03/01/2021] [Indexed: 12/17/2022] Open
Abstract
The SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) is a high-risk viral agent involved in the recent pandemic stated worldwide by the World Health Organization. The infection is correlated to a severe systemic and respiratory disease in many cases, which is clinically treated with a multi-drug pharmacological approach. The purpose of this investigation was to evaluate through a literature overview the effect of adjuvant therapies and supplements for the SARS-CoV-2 infection. The research has analyzed the advantage of the EK1C4, by also assessing the studies on the resveratrol, vitamin D, and melatonin as adjuvant supplements for long hauler patients' prognosis. The evaluated substances reported important benefits for the improvement of the immune system and as a potential inhibitor molecules against SARS-CoV-2, highlighting the use of sartans as therapy. The adjuvant supplements seem to create an advantage for the healing of the long hauler patients affected by chronic symptoms of constant chest and heart pain, intestinal disorders, headache, difficulty concentrating, memory loss, and tachycardia.
Collapse
Affiliation(s)
- Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (C.G.I.); (G.M.); (M.T.D.); (D.H.); (L.S.); (F.I.); (G.D.)
| | - Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (C.G.I.); (G.M.); (M.T.D.); (D.H.); (L.S.); (F.I.); (G.D.)
| | - Ioana Roxana Bordea
- Department of Oral Rehabilitation, Faculty of Dentistry, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (C.G.I.); (G.M.); (M.T.D.); (D.H.); (L.S.); (F.I.); (G.D.)
| | - Edit Xhajanka
- Dental Prosthesis Department, Medical University of Tirana, UMT, Rruga e Dibrës, Tirana 1001, Albania;
| | - Antonio Scarano
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Felice Lorusso
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Marco Farronato
- Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, University of Milan, UOC Maxillo-Facial Surgery and Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, 20100 Milan, Italy; (M.F.); (G.M.T.)
| | - Gianluca Martino Tartaglia
- Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, University of Milan, UOC Maxillo-Facial Surgery and Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, 20100 Milan, Italy; (M.F.); (G.M.T.)
| | - Ciro Gargiulo Isacco
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (C.G.I.); (G.M.); (M.T.D.); (D.H.); (L.S.); (F.I.); (G.D.)
- Human Stem Cells Research Center HSC of Ho Chi Minh, Ho Chi Minh 70000, Vietnam
- Embryology and Regenerative Medicine and Immunology, Pham Chau Trinh University of Medicine Hoi An, Hoi An 70000, Vietnam
| | - Grazia Marinelli
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (C.G.I.); (G.M.); (M.T.D.); (D.H.); (L.S.); (F.I.); (G.D.)
| | - Maria Teresa D’Oria
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (C.G.I.); (G.M.); (M.T.D.); (D.H.); (L.S.); (F.I.); (G.D.)
- Department of Medical and Biological Sciences, Via delle Scienze, Università degli Studi di Udine, 206, 33100 Udine, Italy
| | - Denisa Hazballa
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (C.G.I.); (G.M.); (M.T.D.); (D.H.); (L.S.); (F.I.); (G.D.)
- Kongresi Elbasanit, Rruga: Aqif Pasha, 3001 Elbasan, Albania
| | - Luigi Santacroce
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (C.G.I.); (G.M.); (M.T.D.); (D.H.); (L.S.); (F.I.); (G.D.)
| | - Andrea Ballini
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Campus Universitario, University of Bari, 70125 Bari, Italy;
- Department of Precision Medicine, University of Campania, 80138 Naples, Italy
| | - Maria Contaldo
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Via Luigi de Crecchio, 6, 80138 Naples, Italy;
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (C.G.I.); (G.M.); (M.T.D.); (D.H.); (L.S.); (F.I.); (G.D.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Medicine Aldo Moro, 70124 Bari, Italy; (A.D.I.); (A.M.I.); (C.G.I.); (G.M.); (M.T.D.); (D.H.); (L.S.); (F.I.); (G.D.)
| |
Collapse
|
21
|
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused millions of infections and deaths worldwide since its emergence in December 2019. As there is little or no natural immunity in the human population or specific anti-COVID-19 drugs, researchers from the government, academia and industry are developing vaccines at an unprecedented speed to halt the pandemic. In this review, the results of animal experiments and clinical trials on several vaccine technical platforms are summarized, and several challenges are also discussed to further promote the development, evaluation and application of vaccines during the challenging situation of the global pandemic.
Collapse
Affiliation(s)
- Dan-Dan Li
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, No. 935 Jiaoling Road, Kunming, 650118 Yunnan China
| | - Qi-Han Li
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, No. 935 Jiaoling Road, Kunming, 650118 Yunnan China
| |
Collapse
|
22
|
Gaebler C, Wang Z, Lorenzi JCC, Muecksch F, Finkin S, Tokuyama M, Cho A, Jankovic M, Schaefer-Babajew D, Oliveira TY, Cipolla M, Viant C, Barnes CO, Hurley A, Turroja M, Gordon K, Millard KG, Ramos V, Schmidt F, Weisblum Y, Jha D, Tankelevich M, Yee J, Shimeliovich I, Robbiani DF, Zhao Z, Gazumyan A, Hatziioannou T, Bjorkman PJ, Mehandru S, Bieniasz PD, Caskey M, Nussenzweig MC, Hagglof T, Schwartz RE, Bram Y, Martinez-Delgado G, Mendoza P, Breton G, Dizon J, Unson-O'Brien C, Patel R. Evolution of Antibody Immunity to SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33173867 DOI: 10.1101/2020.11.03.367391] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has infected 78 million individuals and is responsible for over 1.7 million deaths to date. Infection is associated with development of variable levels of antibodies with neutralizing activity that can protect against infection in animal models. Antibody levels decrease with time, but the nature and quality of the memory B cells that would be called upon to produce antibodies upon re-infection has not been examined. Here we report on the humoral memory response in a cohort of 87 individuals assessed at 1.3 and 6.2 months after infection. We find that IgM, and IgG anti-SARS-CoV-2 spike protein receptor binding domain (RBD) antibody titers decrease significantly with IgA being less affected. Concurrently, neutralizing activity in plasma decreases by five-fold in pseudotype virus assays. In contrast, the number of RBD-specific memory B cells is unchanged. Memory B cells display clonal turnover after 6.2 months, and the antibodies they express have greater somatic hypermutation, increased potency and resistance to RBD mutations, indicative of continued evolution of the humoral response. Analysis of intestinal biopsies obtained from asymptomatic individuals 4 months after coronavirus disease-2019 (COVID-19) onset, using immunofluorescence, or polymerase chain reaction, revealed persistence of SARS-CoV-2 nucleic acids and immunoreactivity in the small bowel of 7 out of 14 volunteers. We conclude that the memory B cell response to SARS-CoV-2 evolves between 1.3 and 6.2 months after infection in a manner that is consistent with antigen persistence.
Collapse
|
23
|
[Infectivity-enhancing antibodies against SARS-CoV-2]. Uirusu 2021; 71:169-174. [PMID: 37245979 DOI: 10.2222/jsv.71.169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Antibodies against the receptor binding domain of the spike protein of SARS-CoV-2 play an important role in preventing infection as neutralizing antibodies. However, antibodies that recognize a specific site on the N-terminal domain of the spike protein induce an open domain of receptor binding that increases the binding of ACE2 and enhances the infectivity of SARS-CoV-2. Furthermore, the presence of the infectivity-enhancing antibodies reduces the neutralizing activity of the neutralizing antibodies. Therefore, when considering the antibody response to SARS-CoV-2, it is necessary to consider not only neutralizing antibodies but also the balance between neutralizing and infectivity-enhancing antibodies. In this article, function and mechanism of infectivity-enhancing antibodies are introduced.
Collapse
|
24
|
Winkler ES, Gilchuk P, Yu J, Bailey AL, Chen RE, Zost SJ, Jang H, Huang Y, Allen JD, Case JB, Sutton RE, Carnahan RH, Darling TL, Boon ACM, Mack M, Head RD, Ross TM, Crowe JE, Diamond MS. Human neutralizing antibodies against SARS-CoV-2 require intact Fc effector functions and monocytes for optimal therapeutic protection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 33398272 DOI: 10.1101/2020.12.28.424554] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SARS-CoV-2 has caused the global COVID-19 pandemic. Although passively delivered neutralizing antibodies against SARS-CoV-2 show promise in clinical trials, their mechanism of action in vivo is incompletely understood. Here, we define correlates of protection of neutralizing human monoclonal antibodies (mAbs) in SARS-CoV-2-infected animals. Whereas Fc effector functions are dispensable when representative neutralizing mAbs are administered as prophylaxis, they are required for optimal protection as therapy. When given after infection, intact mAbs reduce SARS-CoV-2 burden and lung disease in mice and hamsters better than loss-of-function Fc variant mAbs. Fc engagement of neutralizing antibodies mitigates inflammation and improves respiratory mechanics, and transcriptional profiling suggests these phenotypes are associated with diminished innate immune signaling and preserved tissue repair. Immune cell depletions establish that neutralizing mAbs require monocytes for therapeutic efficacy. Thus, potently neutralizing mAbs require Fc effector functions for maximal therapeutic benefit during therapy to modulate protective immune responses and mitigate lung disease.
Collapse
|
25
|
Galipeau Y, Greig M, Liu G, Driedger M, Langlois MA. Humoral Responses and Serological Assays in SARS-CoV-2 Infections. Front Immunol 2020; 11:610688. [PMID: 33391281 PMCID: PMC7775512 DOI: 10.3389/fimmu.2020.610688] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/23/2020] [Indexed: 12/14/2022] Open
Abstract
In December 2019, the novel betacoronavirus Severe Acute Respiratory Disease Coronavirus 2 (SARS-CoV-2) was first detected in Wuhan, China. SARS-CoV-2 has since become a pandemic virus resulting in hundreds of thousands of deaths and deep socioeconomic implications worldwide. In recent months, efforts have been directed towards detecting, tracking, and better understanding human humoral responses to SARS-CoV-2 infection. It has become critical to develop robust and reliable serological assays to characterize the abundance, neutralization efficiency, and duration of antibodies in virus-exposed individuals. Here we review the latest knowledge on humoral immune responses to SARS-CoV-2 infection, along with the benefits and limitations of currently available commercial and laboratory-based serological assays. We also highlight important serological considerations, such as antibody expression levels, stability and neutralization dynamics, as well as cross-reactivity and possible immunological back-boosting by seasonal coronaviruses. The ability to accurately detect, measure and characterize the various antibodies specific to SARS-CoV-2 is necessary for vaccine development, manage risk and exposure for healthcare and at-risk workers, and for monitoring reinfections with genetic variants and new strains of the virus. Having a thorough understanding of the benefits and cautions of standardized serological testing at a community level remains critically important in the design and implementation of future vaccination campaigns, epidemiological models of immunity, and public health measures that rely heavily on up-to-date knowledge of transmission dynamics.
Collapse
Affiliation(s)
- Yannick Galipeau
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Matthew Greig
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - George Liu
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | | | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- uOttawa Center for Infection, Immunity and Inflammation (CI3), Ottawa, ON, Canada
| |
Collapse
|