1
|
Cao J, Gan M, Zhang Z, Lin X, Ouyang Q, Fu H, Xu X, Wang Z, Li X, Wang Y, Cai H, Lei Q, Liu L, Wang H, Fan X. A Hidden Guardian: The Stability and Spectrum of Antibody-Dependent Cell-Mediated Cytotoxicity in COVID-19 Response in Chinese Adults. Vaccines (Basel) 2025; 13:262. [PMID: 40266151 PMCID: PMC11945335 DOI: 10.3390/vaccines13030262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 04/24/2025] Open
Abstract
OBJECTIVES Identifying immune-protective biomarkers is crucial for the effective management and mitigation of current and future COVID-19 outbreaks, particularly in preventing or counteracting the immune evasion exhibited by the Omicron variants. The emergence of SARS-CoV-2 variants, especially those within the Omicron lineage, has highlighted their capacity to evade neutralizing antibodies, emphasizing the need to understand the role of antibody-dependent cell-mediated cytotoxicity (ADCC) in combating these infections. METHODS This study, conducted in Qichun City, Hubei province, from December 2021 to March 2023, involved 50 healthy Chinese adults who had received two doses of inactivated vaccines and had subsequently experienced mild infections with the Omicron BA.5 variant. Blood samples from these 50 healthy Chinese adults were collected at six distinct time points: at baseline and at the 1st, 3rd, 6th, and 9th months following the third dose of the inactivated vaccine, as well as 3 months post-breakthrough infection. Their sera were analyzed to assess ADCC and neutralization effects. RESULTS The results indicated that the antibodies elicited by the inactivated SARS-CoV-2 vaccine targeted the spike protein, exhibiting both pre-existing neutralizing and ADCC activities against Omicron variants BA.5 and XBB.1.5. Notably, the ADCC activity demonstrated greater stability compared to that of the neutralizing effects, persisting for at least 15 months post-vaccination, and could be augmented by additional vaccine doses and breakthrough infections. The ADCC effect associated with hybrid immunity effectively targets a spectrum of prospective Omicron variants, including BA.2.86, CH.1.1, EG.5.1, and JN.1. CONCLUSIONS In light of its stability and broad-spectrum efficacy, we recommend the use of the ADCC effect as a biomarker for assessing protective immunity and guiding the development of vaccines and monoclonal antibodies.
Collapse
Affiliation(s)
- Jinge Cao
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan 430030, China; (J.C.); (M.G.); (X.L.); (Q.O.); (H.F.); (X.X.)
| | - Mengze Gan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan 430030, China; (J.C.); (M.G.); (X.L.); (Q.O.); (H.F.); (X.X.)
| | - Zhihao Zhang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.Z.); (Z.W.); (X.L.); (Y.W.); (H.C.); (L.L.)
| | - Xiaosong Lin
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan 430030, China; (J.C.); (M.G.); (X.L.); (Q.O.); (H.F.); (X.X.)
| | - Qi Ouyang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan 430030, China; (J.C.); (M.G.); (X.L.); (Q.O.); (H.F.); (X.X.)
| | - Hui Fu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan 430030, China; (J.C.); (M.G.); (X.L.); (Q.O.); (H.F.); (X.X.)
| | - Xinyue Xu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan 430030, China; (J.C.); (M.G.); (X.L.); (Q.O.); (H.F.); (X.X.)
| | - Zhen Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.Z.); (Z.W.); (X.L.); (Y.W.); (H.C.); (L.L.)
| | - Xinlian Li
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.Z.); (Z.W.); (X.L.); (Y.W.); (H.C.); (L.L.)
| | - Yaxin Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.Z.); (Z.W.); (X.L.); (Y.W.); (H.C.); (L.L.)
| | - Hao Cai
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.Z.); (Z.W.); (X.L.); (Y.W.); (H.C.); (L.L.)
| | - Qing Lei
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Li Liu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.Z.); (Z.W.); (X.L.); (Y.W.); (H.C.); (L.L.)
| | - Hao Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.Z.); (Z.W.); (X.L.); (Y.W.); (H.C.); (L.L.)
| | - Xionglin Fan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan 430030, China; (J.C.); (M.G.); (X.L.); (Q.O.); (H.F.); (X.X.)
| |
Collapse
|
2
|
Mao JH, Chen WM, Wang Y, Wang XM, Shao YH, Liu J, Tu ZC. Serum metabolism-transcriptomics investigated into the immunity of whey protein isolate-galacto-oligosaccharide conjugates after dynamic high-pressure microfluidics pretreatment. Food Res Int 2024; 196:115121. [PMID: 39614521 DOI: 10.1016/j.foodres.2024.115121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/23/2024] [Accepted: 09/19/2024] [Indexed: 12/01/2024]
Abstract
The objective of this study was to investigate the immunomodulatory effects of whey protein isolate (WPI)-galacto-oligosaccharide conjugates following dynamic high-pressure microfluidics pretreatment (DHPM) in cyclophosphamide-induced immunosuppressed mice. DHPM facilitated the conjugation of WPI and galacto-oligosaccharide, and inhibited the generation of fluorescent advanced glycation end products (AGEs) and pentosidine. The conjugates demonstrated a significant immune recovery effect on CTX-induced immunosuppressed mice, as evidenced by the enhancement of IgG antibody levels (from 3.5 to 4.1) and the reduction of the levels of immunosuppressive effector factors TGF-β (from 148.1 to 111.2) and IFN-γ (from 34.4 to 17.9). Furthermore, the conjugates exhibited a notable ability to repair histological lesion in the spleen of CTX-induced immunosuppressed mice. Spleen transcriptomics revealed that the Marco, Klrc3 and Cd209b genes were associated with the immune enhancement activity of the conjugates. Metabolomic analysis identified arginine biosynthesis, sphingolipid metabolism, alanine, aspartate and glutamate metabolism, and phenylalanine, tyrosine and tryptophan biosynthesis as key pathways in the immune enhancement activity of the conjugates. Metabolomics combined with transcriptomics indicated the importance of macrophage activation in the restoration of immunosuppressed mice's immunity by the conjugates. Therefore, the improvement in immunity observed with WPI-galacto-oligosaccharide conjugates may be related to the activation of macrophages.
Collapse
Affiliation(s)
- Ji-Hua Mao
- National R&D Center for Freshwater Fish Processing, College of Life Science, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| | - Wen-Mei Chen
- National R&D Center for Freshwater Fish Processing, College of Life Science, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| | - Yang Wang
- National R&D Center for Freshwater Fish Processing, College of Life Science, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| | - Xu-Mei Wang
- National R&D Center for Freshwater Fish Processing, College of Life Science, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| | - Yan-Hong Shao
- National R&D Center for Freshwater Fish Processing, College of Life Science, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| | - Jun Liu
- National R&D Center for Freshwater Fish Processing, College of Life Science, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China.
| | - Zong-Cai Tu
- National R&D Center for Freshwater Fish Processing, College of Life Science, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China; State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China.
| |
Collapse
|
3
|
Weskamm LM, Tarnow P, Harms C, Huchon M, Raadsen MP, Friedrich M, Rübenacker L, Grüttner C, Garcia MG, Koch T, Becker S, Sutter G, Lhomme E, Haagmans BL, Fathi A, Blois SM, Dahlke C, Richert L, Addo MM. Dissecting humoral immune responses to an MVA-vectored MERS-CoV vaccine in humans using a systems serology approach. iScience 2024; 27:110470. [PMID: 39148710 PMCID: PMC11325358 DOI: 10.1016/j.isci.2024.110470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 06/11/2024] [Accepted: 07/03/2024] [Indexed: 08/17/2024] Open
Abstract
Besides neutralizing antibodies, which are considered an important measure for vaccine immunogenicity, Fc-mediated antibody functions can contribute to antibody-mediated protection. They are strongly influenced by structural antibody properties such as subclass and Fc glycan composition. We here applied a systems serology approach to dissect humoral immune responses induced by MVA-MERS-S, an MVA-vectored vaccine against the Middle East respiratory syndrome coronavirus (MERS-CoV). Building on preceding studies reporting the safety and immunogenicity of MVA-MERS-S, our study highlights the potential of a late boost, administered one year after prime, to enhance both neutralizing and Fc-mediated antibody functionality compared to the primary vaccination series. Distinct characteristics were observed for antibodies specific to the MERS-CoV spike protein S1 and S2 subunits, regarding subclass and glycan compositions as well as Fc functionality. These findings highlight the benefit of a late homologous booster vaccination with MVA-MERS-S and may be of interest for the design of future coronavirus vaccines.
Collapse
Affiliation(s)
- Leonie M Weskamm
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Paulina Tarnow
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Charlotte Harms
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Glyco-HAM, a Cooperation of Universität Hamburg, Technology Platform Mass Spectrometry and University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melanie Huchon
- University of Bordeaux, INSERM, INRIA, BPH, U1219, Sistm, Bordeaux, France
- Vaccine Research Institute, Creteil, France
| | - Matthijs P Raadsen
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Monika Friedrich
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Laura Rübenacker
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Cordula Grüttner
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Mariana G Garcia
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Glyco-HAM, a Cooperation of Universität Hamburg, Technology Platform Mass Spectrometry and University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till Koch
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- Antibiotic Stewardship Team, Pharmacy of the University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, Marburg, Germany
- German Center for Infection Research, Partner Site Gießen-Marburg-Langen, Marburg, Germany
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilian University Munich, Munich, Germany
- German Center for Infection Research, Partner Site München, Munich, Germany
| | - Edouard Lhomme
- University of Bordeaux, INSERM, INRIA, BPH, U1219, Sistm, Bordeaux, France
- Vaccine Research Institute, Creteil, France
- CHU de Bordeaux, Service d'Information Médicale, Bordeaux, France
| | - Bart L Haagmans
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Anahita Fathi
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- Division of Infectious Diseases, 1st Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra M Blois
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Glyco-HAM, a Cooperation of Universität Hamburg, Technology Platform Mass Spectrometry and University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Dahlke
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Laura Richert
- University of Bordeaux, INSERM, INRIA, BPH, U1219, Sistm, Bordeaux, France
- Vaccine Research Institute, Creteil, France
- CHU de Bordeaux, Service d'Information Médicale, Bordeaux, France
| | - Marylyn M Addo
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
4
|
Seki S, Parbie PK, Yamamoto H, Matano T. Virion-surface display of a chimeric immunoglobulin Fc domain facilitating uptake by antigen-presenting cells. J Biotechnol 2024; 391:57-63. [PMID: 38851397 DOI: 10.1016/j.jbiotec.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
Antigen-presenting cells (APCs) play an important role in virus infection control by bridging innate and adaptive immune responses. Macrophages and dendritic cells (DCs) possess various surface receptors to recognize/internalize antigens, and antibody binding can enhance pathogen-opsonizing uptake by these APCs via interaction of antibody fragment crystallizable (Fc) domains with Fc receptors, evoking profound pathogen control in certain settings. Here, we examined phagocytosis-enhancing potential of Fc domains directly oriented on a retroviral virion/virus-like particle (VLP) surface. We generated an expression vector coding a murine Fc fragment fused to the transmembrane region (TM) of a retroviral envelope protein, deriving expression of the Fc-TM fusion protein on the transfected cell surface and production of virions incorporating the chimeric Fc upon co-transfection. Incubation of Fc-displaying simian immunodeficiency virus (SIV) with murine J774 macrophages and bone marrow-derived DCs derived Fc receptor-dependent enhanced uptake, being visualized by imaging cytometry. Alternative preparation of a murine leukemia virus (MLV) backbone-based Fc-displaying VLP loading an influenza virus hemagglutinin (HA) antigen resulted in enhanced HA internalization by macrophages, stating antigen compatibility of the design. Results show that the Fc-TM fusion molecule can be displayed on certain viruses/VLPs and may be utilized as a molecular adjuvant to facilitate APC antigen uptake.
Collapse
Affiliation(s)
- Sayuri Seki
- AIDS Research Center, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Prince Kofi Parbie
- AIDS Research Center, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan; Graduate School of Medical Sciences and Joint Research Center for Human Retrovirus Infection, Kumamoto University, Honjo 2-2-1, Chuo-ku, Kumamoto 860-0811, Japan
| | - Hiroyuki Yamamoto
- Graduate School of Medical Sciences and Joint Research Center for Human Retrovirus Infection, Kumamoto University, Honjo 2-2-1, Chuo-ku, Kumamoto 860-0811, Japan; AIDS Research Center, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashi-Murayama City, Tokyo 208-0011, Japan; Immunodeficiency Laboratory, Department of Biomedicine, University Hospital, 20 Hebelstrasse, Basel 4031, Switzerland.
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan; Graduate School of Medical Sciences and Joint Research Center for Human Retrovirus Infection, Kumamoto University, Honjo 2-2-1, Chuo-ku, Kumamoto 860-0811, Japan; The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| |
Collapse
|
5
|
Beaudoin-Bussières G, Finzi A. Deciphering Fc-effector functions against SARS-CoV-2. Trends Microbiol 2024; 32:756-768. [PMID: 38365562 DOI: 10.1016/j.tim.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 02/18/2024]
Abstract
Major efforts were deployed to study the antibody response against SARS-CoV-2. Antibodies neutralizing SARS-CoV-2 have been extensively studied in the context of infections, vaccinations, and breakthrough infections. Antibodies, however, are pleiotropic proteins that have many functions in addition to neutralization. These include Fc-effector functions such as antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). Although important to combat viral infections, these Fc-effector functions were less studied in the context of SARS-CoV-2 compared with binding and neutralization. This is partly due to the difficulty in developing reliable assays to measure Fc-effector functions compared to antibody binding and neutralization. Multiple assays have now been developed and can be used to measure different Fc-effector functions. Here, we review these assays and what is known regarding anti-SARS-CoV-2 Fc-effector functions. Overall, this review summarizes and updates our current state of knowledge regarding anti-SARS-CoV-2 Fc-effector functions.
Collapse
Affiliation(s)
- Guillaume Beaudoin-Bussières
- Centre de recherche du CHUM, Montréal, Québec H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Québec H2X 0A9, Canada
| | - Andrés Finzi
- Centre de recherche du CHUM, Montréal, Québec H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Québec H2X 0A9, Canada.
| |
Collapse
|
6
|
van der Straten K, Guerra D, Kerster G, Claireaux M, Grobben M, Schriek AI, Boyd A, van Rijswijk J, Tejjani K, Eggink D, Beaumont T, de Taeye SW, de Bree GJ, Sanders RW, van Gils MJ. Primary SARS-CoV-2 variant of concern infections elicit broad antibody Fc-mediated effector functions and memory B cell responses. PLoS Pathog 2024; 20:e1012453. [PMID: 39146376 PMCID: PMC11349224 DOI: 10.1371/journal.ppat.1012453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/27/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024] Open
Abstract
Neutralization of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) by human sera is a strong correlate of protection against symptomatic and severe Coronavirus Disease 2019 (COVID-19). The emergence of antigenically distinct SARS-CoV-2 variants of concern (VOCs) and the relatively rapid waning of serum antibody titers, however, raises questions about the sustainability of serum protection. In addition to serum neutralization, other antibody functionalities and the memory B cell (MBC) response are suggested to help maintaining this protection. In this study, we investigate the breadth of spike (S) protein-specific serum antibodies that mediate effector functions by interacting with Fc-gamma receptor IIa (FcγRIIa) and FcγRIIIa, and of the receptor binding domain (RBD)-specific MBCs, following a primary SARS-CoV-2 infection with the D614G, Alpha, Beta, Gamma, Delta, Omicron BA.1 or BA.2 variant. Irrespectively of the variant causing the infection, the breadth of S protein-specific serum antibodies that interact with FcγRIIa and FcγRIIIa and the RBD-specific MBC responses exceeded the breadth of serum neutralization, although the Alpha-induced B cell response seemed more strain-specific. Between VOC groups, both quantitative and qualitative differences in the immune responses were observed, suggesting differences in immunogenicity. Overall, this study contributes to the understanding of protective humoral and B cell responses in the light of emerging antigenically distinct VOCs, and highlights the need to study the immune system beyond serum neutralization to gain a better understanding of the protection against emerging variants.
Collapse
Affiliation(s)
- Karlijn van der Straten
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Denise Guerra
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Gius Kerster
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Mathieu Claireaux
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Marloes Grobben
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Angela I. Schriek
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Anders Boyd
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Stichting HIV monitoring, Amsterdam, the Netherlands
| | - Jacqueline van Rijswijk
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Khadija Tejjani
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Dirk Eggink
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Tim Beaumont
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Steven W. de Taeye
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Godelieve J. de Bree
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
- Amsterdam UMC, location Academic Medical Center, Department of Internal Medicine, Amsterdam, The Netherlands
| | - Rogier W. Sanders
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Marit J. van Gils
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Wolters RM, Ferguson JA, Nuñez IA, Chen EE, Sornberger T, Myers L, Oeverdieck S, Raghavan SSR, Kona C, Handal LS, Esilu TE, Davidson E, Doranz BJ, Engdahl TB, Kose N, Williamson LE, Creech CB, Gibson-Corley KN, Ward AB, Crowe JE. Isolation of human antibodies against influenza B neuraminidase and mechanisms of protection at the airway interface. Immunity 2024; 57:1413-1427.e9. [PMID: 38823390 PMCID: PMC11440431 DOI: 10.1016/j.immuni.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/16/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024]
Abstract
Influenza B viruses (IBVs) comprise a substantial portion of the circulating seasonal human influenza viruses. Here, we describe the isolation of human monoclonal antibodies (mAbs) that recognized the IBV neuraminidase (NA) glycoprotein from an individual following seasonal vaccination. Competition-binding experiments suggested the antibodies recognized two major antigenic sites. One group, which included mAb FluB-393, broadly inhibited IBV NA sialidase activity, protected prophylactically in vivo, and bound to the lateral corner of NA. The second group contained an active site mAb, FluB-400, that broadly inhibited IBV NA sialidase activity and virus replication in vitro in primary human respiratory epithelial cell cultures and protected against IBV in vivo when administered systemically or intranasally. Overall, the findings described here shape our mechanistic understanding of the human immune response to the IBV NA glycoprotein through the demonstration of two mAb delivery routes for protection against IBV and the identification of potential IBV therapeutic candidates.
Collapse
Affiliation(s)
- Rachael M Wolters
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James A Ferguson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ivette A Nuñez
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Elaine E Chen
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ty Sornberger
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Luke Myers
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Svearike Oeverdieck
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sai Sundar Rajan Raghavan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Chandrahaas Kona
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Laura S Handal
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | - Taylor B Engdahl
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lauren E Williamson
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - C Buddy Creech
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Katherine N Gibson-Corley
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - James E Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
8
|
Gao R, Feng C, Sheng Z, Li F, Wang D. Research progress in Fc-effector functions against SARS-CoV-2. J Med Virol 2024; 96:e29638. [PMID: 38682662 DOI: 10.1002/jmv.29638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/31/2024] [Accepted: 04/18/2024] [Indexed: 05/01/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has caused more than 676 million cases in the global human population with approximately 7 million deaths and vaccination has been proved as the most effective countermeasure in reducing clinical complications and mortality rate of SARS-CoV-2 infection in people. However, the protective elements and correlation of protection induced by vaccination are still not completely understood. Various antibodies with multiple protective mechanisms can be induced simultaneously by vaccination in vivo, thereby complicating the identification and characterization of individual correlate of protection. Recently, an increasing body of observations suggests that antibody-induced Fc-effector functions play a crucial role in combating SARS-CoV-2 infections, including neutralizing antibodies-escaping variants. Here, we review the recent progress in understanding the impact of Fc-effector functions in broadly disarming SARS-CoV-2 infectivity and discuss various efforts in harnessing this conserved antibody function to develop an effective SARS-CoV-2 vaccine that can protect humans against infections by SARS-CoV-2 virus and its variants of concern.
Collapse
Affiliation(s)
- Rongyuan Gao
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Chenchen Feng
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Zizhang Sheng
- Zuckerman Mind Brian Behavior Institute, Columbia University, New York, New York, USA
| | - Feng Li
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, Kentucky, USA
| | - Dan Wang
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
9
|
Zedan HT, Smatti MK, Al-Sadeq DW, Al Khatib HA, Nicolai E, Pieri M, Bernardini S, Hssain AA, Taleb S, Qotba H, Issa K, Abu Raddad LJ, Althani AA, Nasrallah GK, Yassine HM. SARS-CoV-2 infection triggers more potent antibody-dependent cellular cytotoxicity (ADCC) responses than mRNA-, vector-, and inactivated virus-based COVID-19 vaccines. J Med Virol 2024; 96:e29527. [PMID: 38511514 DOI: 10.1002/jmv.29527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 02/08/2024] [Accepted: 03/01/2024] [Indexed: 03/22/2024]
Abstract
Neutralizing antibodies (NAbs) are elicited after infection and vaccination and have been well studied. However, their antibody-dependent cellular cytotoxicity (ADCC) functionality is still poorly characterized. Here, we investigated ADCC activity in convalescent sera from infected patients with wild-type (WT) severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) or omicron variant compared with three coronavirus disease 2019 (COVID-19) vaccine platforms and postvaccination breakthrough infection (BTI). We analyzed ADCC activity targeting SARS-CoV-2 spike (S) and nucleocapsid (N) proteins in convalescent sera following WT SARS-CoV-2-infection (n = 91), including symptomatic and asymptomatic infections, omicron-infection (n = 8), COVID-19 vaccination with messenger RNA- (mRNA)- (BNT162b2 or mRNA-1273, n = 77), adenovirus vector- (n = 41), and inactivated virus- (n = 46) based vaccines, as well as post-mRNA vaccination BTI caused by omicron (n = 28). Correlations between ADCC, binding, and NAb titers were reported. ADCC was elicited within the first month postinfection and -vaccination and remained detectable for ≥3 months. WT-infected symptomatic patients had higher S-specific ADCC levels than asymptomatic and vaccinated individuals. Also, no difference in N-specific ADCC activity was seen between symptomatic and asymptomatic patients, but the levels were higher than the inactivated vaccine. Notably, omicron infection showed reduced overall ADCC activity compared to WT SARS-CoV-2 infection. Although post-mRNA vaccination BTI elicited high levels of binding and NAbs, ADCC activity was significantly reduced. Also, there was no difference in ADCC levels across the four vaccines, although NAbs and binding antibody titers were significantly higher in mRNA-vaccinated individuals. All evaluated vaccine platforms are inferior in inducing ADCC compared to natural infection with WT SARS-CoV-2. The inactivated virus-based vaccine can induce N-specific ADCC activity, but its relevance to clinical outcomes requires further investigation. Our data suggest that ADCC could be used to estimate the extra-neutralization level against COVID-19 and provides evidence that vaccination should focus on other Fc-effector functions besides NAbs. Also, the decreased susceptibility of the omicron variant to ADCC offers valuable guidance for forthcoming efforts to identify the specific targets of antibodies facilitating ADCC.
Collapse
Affiliation(s)
- Hadeel T Zedan
- Infectious Diseases Department, Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, Doha, Qatar
| | - Maria K Smatti
- Infectious Diseases Department, Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
| | - Duaa W Al-Sadeq
- College of Medicine, Member of QU Health, Qatar University, Doha, Qatar
| | - Hebah A Al Khatib
- Infectious Diseases Department, Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
| | - Eleonora Nicolai
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Massimo Pieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ali Ait Hssain
- Medical Intensive Care Unit, Hamad Medical Corporation, Doha, Qatar
| | - Sara Taleb
- Department of Research, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Hamda Qotba
- Department of Clinical Research, Primary Health Care Centers, Doha, Qatar
- Department of Pathology, Sidra Medicine, Doha, Qatar
| | - Khodr Issa
- Proteomics, Inflammatory Response, and Mass Spectrometry (PRISM) Laboratory, INSERM U-1192, University of Lille, Lille, France
| | - Laith J Abu Raddad
- Department of Population Health Sciences, Infectious Disease Epidemiology Group, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Asmaa A Althani
- Infectious Diseases Department, Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, Doha, Qatar
| | - Gheyath K Nasrallah
- Infectious Diseases Department, Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, Doha, Qatar
| | - Hadi M Yassine
- Infectious Diseases Department, Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
| |
Collapse
|
10
|
Wu YH, Chang WT, Hsu CL, Lu YF, Wang JT, Tzeng SJ. FcγRIIB modulates splenic germinal center response against immune subversion during acute influenza A virus infection. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2024; 57:64-75. [PMID: 38087748 DOI: 10.1016/j.jmii.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/02/2023] [Accepted: 11/28/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND B cells are essential for providing humoral protection against acute influenza A virus (IAV) infection. FcγRIIB, a regulator of antibody (Ab) production, influences immune responses during pathogen infections, but its specific impact on humoral protection and B cell-mediated responses against IAV remains unclear. METHODS To investigate FcγRIIB's role in host defense and B cell function during acute IAV infection, we generated mice with systemic FcγRIIB deficiency, functional impairment, and B cell-specific FcγRIIB deletion. We infected these mice with PR8 (H1N1) or Hkx31 (H3N2) IAVs and evaluated body weight preservation, survival rates, Ab production, viral neutralization, Ab affinity maturation, and germinal center B cell development. RESULTS Mice lacking FcγRIIB or with impaired function showed improved protection, preserved body weight, and increased survival rates during IAV infection. Notably, mice with haploinsufficient FcγRIIB function displayed protective effects. Selective deficiency of FcγRIIB in B cells led to enhanced Ab production, resulting in elevated IAV-specific Abs in the serum with superior viral neutralizing potency. However, the impact on the affinity maturation index of virus-specific Abs was modest. Accordingly, FcγRIIB-deficient B cells maintained normal germinal center B cell development during IAV infection, whereas wild-type mice exhibited delayed differentiation. CONCLUSION Our research underscores the pivotal role of FcγRIIB in host defense and B cell-mediated immunity during acute IAV infection. Additionally, our discoveries hold implications for antiviral treatments, particularly during the initial stages of IAV infection, aimed at enhancing the host's humoral immune response.
Collapse
Affiliation(s)
- Yu-Hsuan Wu
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taiwan.
| | - Wan-Ting Chang
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taiwan.
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taiwan.
| | - Yan-Fong Lu
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taiwan; Department of Obstetrics and Gynecology, Shin-Kong Wu Ho-Su Memorial Hospital, Taiwan.
| | - Jann-Tay Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taiwan.
| | - Shiang-Jong Tzeng
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taiwan.
| |
Collapse
|
11
|
Flores A, Alonso-Vega C, Hermann E, Torrico MC, Montaño Villarroel NA, Torrico F, Carlier Y, Truyens C. Monocytes from Uninfected Neonates Born to Trypanosoma cruzi-Infected Mothers Display Upregulated Capacity to Produce TNF-α and to Control Infection in Association with Maternally Transferred Antibodies. Pathogens 2023; 12:1103. [PMID: 37764911 PMCID: PMC10536721 DOI: 10.3390/pathogens12091103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Activated monocytes/macrophages that produce inflammatory cytokines and nitric oxide are crucial for controlling Trypanosoma cruzi infection. We previously showed that uninfected newborns from T. cruzi infected mothers (M+B- newborns) were sensitized to produce higher levels of inflammatory cytokines than newborns from uninfected mothers (M-B- newborns), suggesting that their monocytes were more activated. Thus, we wondered whether these cells might help limit congenital infection. We investigated this possibility by studying the activation status of M+B- cord blood monocytes and their ability to control T. cruzi in vitro infection. We showed that M+B- monocytes have an upregulated capacity to produce the inflammatory cytokine TNF-α and a better ability to control T. cruzi infection than M-B- monocytes. Our study also showed that T. cruzi-specific Abs transferred from the mother play a dual role by favoring trypomastigote entry into M+B- monocytes and inhibiting intracellular amastigote multiplication. These results support the possibility that some M+B- fetuses may eliminate the parasite transmitted in utero from their mothers, thus being uninfected at birth.
Collapse
Affiliation(s)
- Amilcar Flores
- Facultad de Medicina, Universidad Mayor de San Simon (U.M.S.S.), Cochabamba 2500, Bolivia
| | - Cristina Alonso-Vega
- Facultad de Medicina, Universidad Mayor de San Simon (U.M.S.S.), Cochabamba 2500, Bolivia
| | - Emmanuel Hermann
- Laboratory of Parasitology, Faculty of Medicine, ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (U.L.B.), 1070 Brussels, Belgium
| | - Mary-Cruz Torrico
- Facultad de Medicina, Universidad Mayor de San Simon (U.M.S.S.), Cochabamba 2500, Bolivia
| | | | - Faustino Torrico
- Facultad de Medicina, Universidad Mayor de San Simon (U.M.S.S.), Cochabamba 2500, Bolivia
| | - Yves Carlier
- Laboratory of Parasitology, Faculty of Medicine, ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (U.L.B.), 1070 Brussels, Belgium
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70118, USA
| | - Carine Truyens
- Laboratory of Parasitology, Faculty of Medicine, ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (U.L.B.), 1070 Brussels, Belgium
| |
Collapse
|
12
|
Azulay A, Cohen-Lavi L, Friedman LM, McGargill MA, Hertz T. Mapping antibody footprints using binding profiles. CELL REPORTS METHODS 2023; 3:100566. [PMID: 37671022 PMCID: PMC10475849 DOI: 10.1016/j.crmeth.2023.100566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023]
Abstract
The increasing use of monoclonal antibodies (mAbs) in biology and medicine necessitates efficient methods for characterizing their binding epitopes. Here, we developed a high-throughput antibody footprinting method based on binding profiles. We used an antigen microarray to profile 23 human anti-influenza hemagglutinin (HA) mAbs using HA proteins of 43 human influenza strains isolated between 1918 and 2018. We showed that the mAb's binding profile can be used to characterize its influenza subtype specificity, binding region, and binding site. We present mAb-Patch-an epitope prediction method that is based on a mAb's binding profile and the 3D structure of its antigen. mAb-Patch was evaluated using four mAbs with known solved mAb-HA structures. mAb-Patch identifies over 67% of the true epitope when considering only 50-60 positions along the antigen. Our work provides proof of concept for utilizing antibody binding profiles to screen large panels of mAbs and to down-select antibodies for further functional studies.
Collapse
Affiliation(s)
- Asaf Azulay
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- National Institute of Biotechnology in the Negev, Beer-Sheva, Israel
| | - Liel Cohen-Lavi
- National Institute of Biotechnology in the Negev, Beer-Sheva, Israel
- Department of Industrial Engineering and Management, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Lilach M. Friedman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- National Institute of Biotechnology in the Negev, Beer-Sheva, Israel
| | - Maureen A. McGargill
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Tomer Hertz
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- National Institute of Biotechnology in the Negev, Beer-Sheva, Israel
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
13
|
Masson LC, Servian CDP, Jardim VH, Dos Anjos D, Dorta ML, Batalha-Carvalho JV, Moro AM, Romão PRT, Souza M, Fiaccadori FS, Fonseca SG. Heterologous Booster with BNT162b2 Induced High Specific Antibody Levels in CoronaVac Vaccinees. Vaccines (Basel) 2023; 11:1183. [PMID: 37514999 PMCID: PMC10383528 DOI: 10.3390/vaccines11071183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/21/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
Immune responses after COVID-19 vaccination should be evaluated in different populations around the world. This study compared antibody responses induced by ChAdOx1 nCoV-19, CoronaVac, and BNT162b2 vaccines. Blood samples from vaccinees were collected pre- and post-vaccinations with the second and third doses. The study enrolled 78 vaccinees, of whom 62.8% were women, with the following median ages: 26 years-ChAdOx1 nCoV-19; 40 years-CoronaVac; 30 years-BNT162b2. Serum samples were quantified for anti-RBD IgG and anti-RBD IgA and anti-spike IgG by ELISA. After two vaccine doses, BNT162b2 vaccinees produced higher levels of anti-RBD IgA and IgG, and anti-spike IgG compared to ChAdOx1 nCoV-19 and CoronaVac vaccinees. The third dose booster with BNT162b2 induced higher levels of anti-RBD IgA and IgG, and anti-spike IgG in CoronaVac vaccinees. Individuals who reported a SARS-CoV-2 infection before or during the study had higher anti-RBD IgA and IgG production. In conclusion, two doses of the studied vaccines induced detectable levels of anti-RBD IgA and IgG and anti-spike IgG in vaccinees. The heterologous booster with BNT162b2 increased anti-RBD IgA and IgG and anti-spike IgG levels in CoronaVac vaccinees and anti-RBD IgA levels in ChAdOx1 nCoV-19 vaccinees. Furthermore, SARS-CoV-2 infection induced higher anti-RBD IgA and IgG levels in CoronaVac vaccinees.
Collapse
Affiliation(s)
- Letícia Carrijo Masson
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Carolina do Prado Servian
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Vitor Hugo Jardim
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Déborah Dos Anjos
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Miriam Leandro Dorta
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | | | - Ana Maria Moro
- Laboratório de Biofármacos, Instituto Butantan, São Paulo 05503-900, SP, Brazil
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (iii-INCT), Goiânia 74605-050, GO, Brazil
| | - Pedro Roosevelt Torres Romão
- Laboratório de Imunologia Celular e Molecular, Programa de Pós-Graduação em Ciências da Saúde, Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre 90050-170, RS, Brazil
| | - Menira Souza
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Fabiola Souza Fiaccadori
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Simone Gonçalves Fonseca
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (iii-INCT), Goiânia 74605-050, GO, Brazil
| |
Collapse
|
14
|
Purcell RA, Theisen RM, Arnold KB, Chung AW, Selva KJ. Polyfunctional antibodies: a path towards precision vaccines for vulnerable populations. Front Immunol 2023; 14:1183727. [PMID: 37600816 PMCID: PMC10433199 DOI: 10.3389/fimmu.2023.1183727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 08/22/2023] Open
Abstract
Vaccine efficacy determined within the controlled environment of a clinical trial is usually substantially greater than real-world vaccine effectiveness. Typically, this results from reduced protection of immunologically vulnerable populations, such as children, elderly individuals and people with chronic comorbidities. Consequently, these high-risk groups are frequently recommended tailored immunisation schedules to boost responses. In addition, diverse groups of healthy adults may also be variably protected by the same vaccine regimen. Current population-based vaccination strategies that consider basic clinical parameters offer a glimpse into what may be achievable if more nuanced aspects of the immune response are considered in vaccine design. To date, vaccine development has been largely empirical. However, next-generation approaches require more rational strategies. We foresee a generation of precision vaccines that consider the mechanistic basis of vaccine response variations associated with both immunogenetic and baseline health differences. Recent efforts have highlighted the importance of balanced and diverse extra-neutralising antibody functions for vaccine-induced protection. However, in immunologically vulnerable populations, significant modulation of polyfunctional antibody responses that mediate both neutralisation and effector functions has been observed. Here, we review the current understanding of key genetic and inflammatory modulators of antibody polyfunctionality that affect vaccination outcomes and consider how this knowledge may be harnessed to tailor vaccine design for improved public health.
Collapse
Affiliation(s)
- Ruth A. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Robert M. Theisen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
15
|
Cruz Amaya J, Walcheck B, Smith-Gagen J, Lombardi VC, Hudig D. Detection of Antibody-Dependent Cell-Mediated Cytotoxicity-Supporting Antibodies by NK-92-CD16A Cell Externalization of CD107a: Recognition of Antibody Afucosylation and Assay Optimization. Antibodies (Basel) 2023; 12:44. [PMID: 37489366 PMCID: PMC10366760 DOI: 10.3390/antib12030044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/26/2023] Open
Abstract
Antibody-dependent cell-mediated cytotoxicity (ADCC) by natural killer (NK) lymphocytes eliminates cells infected with viruses. Anti-viral ADCC requires three components: (1) antibody; (2) effector lymphocytes with the Fc-IgG receptor CD16A; and (3) viral proteins in infected cell membranes. Fc-afucosylated antibodies bind with greater affinity to CD16A than fucosylated antibodies; individuals' variation in afucosylation contributes to differences in ADCC. Current assays for afucosylated antibodies involve expensive methods. We report an improved bioassay for antibodies that supports ADCC, which encompasses afucosylation. This assay utilizes the externalization of CD107a by NK-92-CD16A cells after antibody recognition. We used anti-CD20 monoclonal antibodies, GA101 WT or glycoengineered (GE), 10% or ~50% afucosylated, and CD20-positive Raji target cells. CD107a increased detection 7-fold compared to flow cytometry to detect Raji-bound antibodies. WT and GE antibody effective concentrations (EC50s) for CD107a externalization differed by 20-fold, with afucosylated GA101-GE more detectable. The EC50s for CD107a externalization vs. 51Cr cell death were similar for NK-92-CD16A and blood NK cells. Notably, the % CD107a-positive cells were negatively correlated with dead Raji cells and were nearly undetectable at high NK:Raji ratios required for cytotoxicity. This bioassay is very sensitive and adaptable to assess anti-viral antibodies but unsuitable as a surrogate assay to monitor cell death after ADCC.
Collapse
Affiliation(s)
- Judith Cruz Amaya
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| | - Bruce Walcheck
- Department of Veterinary and Biological Sciences, Center for Immunology and Masonic Cancer Center, University of Minnesota, 295J AS/VM Building, 1988 Fitch Avenue, Saint Paul, MN 55108, USA
| | - Julie Smith-Gagen
- School of Community Health Sciences, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| | - Vincent C. Lombardi
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| | - Dorothy Hudig
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| |
Collapse
|
16
|
Struble EB, Rawson JMO, Stantchev T, Scott D, Shapiro MA. Uses and Challenges of Antiviral Polyclonal and Monoclonal Antibody Therapies. Pharmaceutics 2023; 15:pharmaceutics15051538. [PMID: 37242780 DOI: 10.3390/pharmaceutics15051538] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Viral diseases represent a major public health concerns and ever-present risks for developing into future pandemics. Antiviral antibody therapeutics, either alone or in combination with other therapies, emerged as valuable preventative and treatment options, including during global emergencies. Here we will discuss polyclonal and monoclonal antiviral antibody therapies, focusing on the unique biochemical and physiological properties that make them well-suited as therapeutic agents. We will describe the methods of antibody characterization and potency assessment throughout development, highlighting similarities and differences between polyclonal and monoclonal products as appropriate. In addition, we will consider the benefits and challenges of antiviral antibodies when used in combination with other antibodies or other types of antiviral therapeutics. Lastly, we will discuss novel approaches to the characterization and development of antiviral antibodies and identify areas that would benefit from additional research.
Collapse
Affiliation(s)
- Evi B Struble
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics CMC, Office of Therapeutic Products, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Jonathan M O Rawson
- Division of Antivirals, Office of Infectious Diseases, Office of New Drugs, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Tzanko Stantchev
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Dorothy Scott
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics CMC, Office of Therapeutic Products, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Marjorie A Shapiro
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
17
|
Loos C, Coccia M, Didierlaurent AM, Essaghir A, Fallon JK, Lauffenburger D, Luedemann C, Michell A, van der Most R, Zhu AL, Alter G, Burny W. Systems serology-based comparison of antibody effector functions induced by adjuvanted vaccines to guide vaccine design. NPJ Vaccines 2023; 8:34. [PMID: 36890168 PMCID: PMC9992919 DOI: 10.1038/s41541-023-00613-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 01/27/2023] [Indexed: 03/10/2023] Open
Abstract
The mechanisms by which antibodies confer protection vary across vaccines, ranging from simple neutralization to functions requiring innate immune recruitment via Fc-dependent mechanisms. The role of adjuvants in shaping the maturation of antibody-effector functions remains under investigated. Using systems serology, we compared adjuvants in licensed vaccines (AS01B/AS01E/AS03/AS04/Alum) combined with a model antigen. Antigen-naive adults received two adjuvanted immunizations followed by late revaccination with fractional-dosed non-adjuvanted antigen ( NCT00805389 ). A dichotomy in response quantities/qualities emerged post-dose 2 between AS01B/AS01E/AS03 and AS04/Alum, based on four features related to immunoglobulin titers or Fc-effector functions. AS01B/E and AS03 induced similar robust responses that were boosted upon revaccination, suggesting that memory B-cell programming by the adjuvanted vaccinations dictated responses post non-adjuvanted boost. AS04 and Alum induced weaker responses, that were dissimilar with enhanced functionalities for AS04. Distinct adjuvant classes can be leveraged to tune antibody-effector functions, where selective vaccine formulation using adjuvants with different immunological properties may direct antigen-specific antibody functions.
Collapse
Affiliation(s)
- Carolin Loos
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | | | - Arnaud M Didierlaurent
- GSK, Rixensart, Belgium.,Center of Vaccinology, University of Geneva, Geneva, Switzerland
| | | | | | | | | | - Ashlin Michell
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | | | - Alex Lee Zhu
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.,Virology and Immunology Program, University of Duisburg-Essen, Essen, Germany
| | - Galit Alter
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | | |
Collapse
|
18
|
Wang W, Yu C, Cui Y, Liu C, Yang Y, Xu G, Wu G, Du J, Fu Z, Guo L, Long C, Xia X, Li Y, Wang L, Wang Y. Development of a reporter gene assay for antibody dependent cellular cytotoxicity activity determination of anti-rabies virus glycoprotein antibodies. Microbiol Immunol 2023; 67:69-78. [PMID: 36346082 DOI: 10.1111/1348-0421.13036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/09/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022]
Abstract
Rabies is a viral disease that is nearly 100% fatal once clinical signs and symptoms develop. Post-exposure prophylaxis can efficiently prevent rabies, and antibody (Ab) induction by vaccination or passive immunization of human rabies immunoglobulin (HRIG) or monoclonal antibodies (mAbs) play an integral role in prevention against rabies. In addition to their capacity to neutralize viruses, antibodies exert their antiviral effects by antibody-dependent cellular cytotoxicity (ADCC), which plays an important role in antiviral immunity and clearance of viral infections. For antibodies against rabies virus (RABV), evaluation of ADCC activity was neglected. Here, we developed a robust cell-based reporter gene assay (RGA) for the determination of the ADCC activity of anti-RABV antibodies using CVS-N2c-293 cells, which stably express the glycoprotein (G) of RABV strain CVS-N2c as target cells, and Jurkat cells, which stably express FcγRⅢa and nuclear factor of activated T cells (NFAT) reporter gene as effector cells (Jurkat/NFAT-luc/FcγRⅢa cells). The experimental parameters were carefully optimized, and the established ADCC assay was systematically validated according to the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) Q2 guideline. We also evaluated the ADCC activity of anti-RABV antibodies, including mAbs, HRIG, and vaccine induced antisera, and found that all test antibodies exhibited ADCC activity with varied strengths. The established RGA provides a novel method for evaluating the ADCC of anti-RABV antibodies.
Collapse
Affiliation(s)
- Wenbo Wang
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Chuanfei Yu
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Yongfei Cui
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Chunyu Liu
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Yalan Yang
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Gangling Xu
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Gang Wu
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Jialiang Du
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Zhihao Fu
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Luyong Guo
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Caifeng Long
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Xijie Xia
- China Pharmaceutical University, Nanjing, China
| | - Yuhua Li
- Division of Arboviral Vaccine, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Lan Wang
- Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| |
Collapse
|
19
|
Batty CJ, Lifshits LM, Hendy DA, Eckshtain-Levi M, Ontiveros-Padilla LA, Carlock MA, Ross TM, Bachelder EM, Ainslie KM. Vinyl Sulfone-functionalized Acetalated Dextran Microparticles as a Subunit Broadly Acting Influenza Vaccine. AAPS J 2023; 25:22. [PMID: 36720729 DOI: 10.1208/s12248-023-00786-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 01/18/2023] [Indexed: 02/02/2023] Open
Abstract
Influenza is a global health concern with millions of infections occurring yearly. Seasonal flu vaccines are one way to combat this virus; however, they are poorly protective against influenza as the virus is constantly mutating, particularly at the immunodominant hemagglutinin (HA) head group. A more broadly acting approach involves Computationally Optimized Broadly Reactive Antigen (COBRA). COBRA HA generates a broad immune response that is capable of protecting against mutating strains. Unfortunately, protein-based vaccines are often weekly immunogenic, so to help boost the immune response, we employed the use of acetalated dextran (Ace-DEX) microparticles (MPs) two ways: one to conjugate COBRA HA to the surface and a second to encapsulate cGAMP. To conjugate the COBRA HA to the surface of the Ace-DEX MPs, a poly(L-lactide)-polyethylene glycol co-polymer with a vinyl sulfone terminal group (PLLA-PEG-VS) was used. MPs encapsulating the STING agonist cGAMP were co-delivered with the antigen to form a broadly active influenza vaccine. This vaccine approach was evaluated in vivo with a prime-boost-boost vaccination schedule and illustrated generation of a humoral and cellular response that could protect against a lethal challenge of A/California/07/2009 in BALB/c mice.
Collapse
Affiliation(s)
- Cole J Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Liubov M Lifshits
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Dylan A Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Meital Eckshtain-Levi
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Luis A Ontiveros-Padilla
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Michael A Carlock
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, Florida, USA
| | - Ted M Ross
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, Florida, USA.,Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA.,Department of Infectious Diseases, University of Georgia, Athens, Goergia, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA. .,Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA. .,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, USA.
| |
Collapse
|
20
|
Voronina DV, Shcheblyakov DV, Favorskaya IA, Esmagambetov IB, Dzharullaeva AS, Tukhvatulin AI, Zubkova OV, Popova O, Kan VY, Bandelyuk AS, Shmarov MM, Logunov DY, Naroditskiy BS, Gintsburg AL. Cross-Reactive Fc-Fused Single-Domain Antibodies to Hemagglutinin Stem Region Protect Mice from Group 1 Influenza a Virus Infection. Viruses 2022; 14:v14112485. [PMID: 36366583 PMCID: PMC9698552 DOI: 10.3390/v14112485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022] Open
Abstract
The continued evolution of influenza viruses reduces the effectiveness of vaccination and antiviral drugs. The identification of novel and universal agents for influenza prophylaxis and treatment is an urgent need. We have previously described two potent single-domain antibodies (VHH), G2.3 and H1.2, which bind to the stem domain of hemagglutinin and efficiently neutralize H1N1 and H5N2 influenza viruses in vivo. In this study, we modified these VHHs with Fc-fragment to enhance their antiviral activity. Reformatting of G2.3 into bivalent Fc-fusion molecule increased its in vitro neutralizing activity against H1N1 and H2N3 viruses up to 80-fold and, moreover, resulted in obtaining the ability to neutralize H5N2 and H9N2 subtypes. We demonstrated that a dose as low as 0.6 mg/kg of G2.3-Fc or H1.2-Fc administered systemically or locally before infection could protect mice from lethal challenges with both H1N1 and H5N2 viruses. Furthermore, G2.3-Fc reduced the lung viral load to an undetectable level. Both VHH-Fc antibodies showed in vivo therapeutic efficacy when delivered via systemic or local route. The findings support G2.3-Fc as a potential therapeutic agent for both prophylaxis and therapy of Group 1 influenza A infection.
Collapse
Affiliation(s)
- Daria V. Voronina
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
- Correspondence:
| | - Dmitry V. Shcheblyakov
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Irina A. Favorskaya
- Medical Microbiology Department, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Ilias B. Esmagambetov
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Alina S. Dzharullaeva
- Medical Microbiology Department, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Amir I. Tukhvatulin
- Medical Microbiology Department, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Olga V. Zubkova
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Olga Popova
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Vladislav Y. Kan
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Alina S. Bandelyuk
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Maxim M. Shmarov
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Denis Y. Logunov
- Medical Microbiology Department, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Boris S. Naroditskiy
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Aleksandr L. Gintsburg
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| |
Collapse
|
21
|
George PJ, Marches R, Nehar-Belaid D, Banchereau J, Lustigman S. The Th1/Tfh-like biased responses elicited by the rASP-1 innate adjuvant are dependent on TRIF and Type I IFN receptor pathways. Front Immunol 2022; 13:961094. [PMID: 36119026 PMCID: PMC9478378 DOI: 10.3389/fimmu.2022.961094] [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: 06/03/2022] [Accepted: 08/12/2022] [Indexed: 11/23/2022] Open
Abstract
Ov-ASP-1 (rASP-1), a parasite-derived protein secreted by the helminth Onchocerca volvulus, is an adjuvant which enhances the potency of the influenza trivalent vaccine (IIV3), even when used with 40-fold less IIV3. This study is aimed to provide a deeper insight into the molecular networks that underline the adjuvanticity of rASP-1. Here we show that rASP-1 stimulates mouse CD11c+ bone marrow-derived dendritic (BMDCs) to secrete elevated levels of IL-12p40, TNF-α, IP-10 and IFN-β in a TRIF-dependent but MyD88-independent manner. rASP-1-activated BMDCs promoted the differentiation of naïve CD4+ T cells into Th1 cells (IFN-γ+) that was TRIF- and type I interferon receptor (IFNAR)-dependent, and into Tfh-like cells (IL21+) and Tfh1 (IFN-γ+ IL21+) that were TRIF-, MyD88- and IFNAR-dependent. rASP-1-activated BMDCs promoted the differentiation of naïve CD4+ T cells into Th17 (IL-17+) cells only when the MyD88 pathway was inhibited. Importantly, rASP-1-activated human blood cDCs expressed upregulated genes that are associated with DC maturation, type I IFN and type II IFN signaling, as well as TLR4-TRIF dependent signaling. These activated cDCs promoted the differentiation of naïve human CD4+ T cells into Th1, Tfh-like and Th17 cells. Our data thus confirms that the rASP-1 is a potent innate adjuvant that polarizes the adaptive T cell responses to Th1/Tfh1 in both mouse and human DCs. Notably, the rASP-1-adjuvanted IIV3 vaccine elicited protection of mice from a lethal H1N1 infection that is also dependent on the TLR4-TRIF axis and IFNAR signaling pathway, as well as on its ability to induce anti-IIV3 antibody production.
Collapse
Affiliation(s)
- Parakkal Jovvian George
- Laboratory Molecular Parasitology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| | - Radu Marches
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | | | - Jacques Banchereau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | - Sara Lustigman
- Laboratory Molecular Parasitology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| |
Collapse
|
22
|
O’Donnell JS, Isaacs A, Jakob V, Lebas C, Barnes JB, Reading PC, Young PR, Watterson D, Dubois PM, Collin N, Chappell KJ. Characterization and comparison of novel adjuvants for a prefusion clamped MERS vaccine. Front Immunol 2022; 13:976968. [PMID: 36119058 PMCID: PMC9478912 DOI: 10.3389/fimmu.2022.976968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Various chemical adjuvants are available to augment immune responses to non-replicative, subunit vaccines. Optimized adjuvant selection can ensure that vaccine-induced immune responses protect against the diversity of pathogen-associated infection routes, mechanisms of infectious spread, and pathways of immune evasion. In this study, we compare the immune response of mice to a subunit vaccine of Middle Eastern respiratory syndrome coronavirus (MERS-CoV) spike protein, stabilized in its prefusion conformation by a proprietary molecular clamp (MERS SClamp) alone or formulated with one of six adjuvants: either (i) aluminium hydroxide, (ii) SWE, a squalene-in-water emulsion, (iii) SQ, a squalene-in-water emulsion containing QS21 saponin, (iv) SMQ, a squalene-in-water emulsion containing QS21 and a synthetic toll-like receptor 4 (TLR4) agonist 3D-6-acyl Phosphorylated HexaAcyl Disaccharide (3D6AP); (v) LQ, neutral liposomes containing cholesterol, 1.2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) and QS21, (vi) or LMQ, neutral liposomes containing cholesterol, DOPC, QS21, and 3D6AP. All adjuvanted formulations induced elevated antibody titers which where greatest for QS21-containing formulations. These had elevated neutralization capacity and induced higher frequencies of IFNƔ and IL-2-producing CD4+ and CD8+ T cells. Additionally, LMQ-containing formulations skewed the antibody response towards IgG2b/c isotypes, allowing for antibody-dependent cellular cytotoxicity. This study highlights the utility of side-by-side adjuvant comparisons in vaccine development.
Collapse
Affiliation(s)
- Jake S. O’Donnell
- The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Ariel Isaacs
- The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | | | - Celia Lebas
- Vaccine Formulation Institute, Geneva, Switzerland
| | - James B. Barnes
- The WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Patrick C. Reading
- The WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Paul R. Young
- The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Daniel Watterson
- The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | | | - Nicolas Collin
- Vaccine Formulation Institute, Geneva, Switzerland
- *Correspondence: Keith J. Chappell, ; Nicolas Collin,
| | - Keith J. Chappell
- The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, QLD, Australia
- *Correspondence: Keith J. Chappell, ; Nicolas Collin,
| |
Collapse
|
23
|
Hu G, Jin WP, Yang ZH, Lv SY, Wu J, Yu YT, Meng SL, Guo J, Wang ZJ, Shen S. Efficacy of Coxsackievirus A2 vaccine candidates correlating to humoral immunity in mice challenged with a mouse-adapted strain. Vaccine 2022; 40:4716-4725. [PMID: 35760737 DOI: 10.1016/j.vaccine.2022.06.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 05/06/2022] [Accepted: 06/05/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND In recent years, Coxsackievirus A2 (CV-A2) has become one of the main serotypes of enterovirus species A associated with hand, foot and mouth disease (HFMD) in China. It has also caused HFMD epidemics in many countries all over the world. Currently, there are no effective, preventive vaccines against it. METHODS A CV-A2 strain was isolated in RD cells and then adapted to grow in Vero cells. This is in compliance with guidelines for cell substrates allowed for human vaccines by the Chinese regulatory authority. Groups of newborn Kunming mice were inoculated on day 3 and day 9 using two formulations of candidate vaccines, empty particles and full particles. They were then challenged on day 14 at a lethal dose with a mouse-adapted strain. RESULTS The mice in the control group all died within 14 days post-challenge whereas most of the mice in the candidate vaccine groups survived. It was found that the titers of neutralizing antibodies was dose-dependent in sera of immunized mice. The results also showed that the vaccine candidates stimulated a strong humoral immune response and protected the mice from disease and death. The virus loads in tissues or organs were significantly reduced and pathological changes were either weak or not observed in the immunized groups compared with those in Al(OH)3 control group. Preliminary mapping of the nucleotide and amino acid residues potentially related to cell tropism of the vaccine strain and virulence of the challenge strain was performed. CONCLUSION The results showed that the RD cell-isolated and Vero cell-adapted CV-A2 strain is a promising vaccine candidate. This active immunization-challenge mouse model mimics the vaccination and then exposure to wildtype viruses, compared with passive immunization-challenge model, and is invaluable for efficacy evaluation in studies on multivalent vaccines containing CV-A2 against HFMD.
Collapse
Affiliation(s)
- Gang Hu
- Wuhan Institute of Biological Products Co. Ltd., No.1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430207, China
| | - Wei-Ping Jin
- Wuhan Institute of Biological Products Co. Ltd., No.1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430207, China
| | - Zhi-Hui Yang
- Wuhan Institute of Biological Products Co. Ltd., No.1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430207, China
| | - Shi-Yun Lv
- Wuhan Institute of Biological Products Co. Ltd., No.1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430207, China
| | - Jie Wu
- Wuhan Institute of Biological Products Co. Ltd., No.1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430207, China
| | - Yu-Ting Yu
- Wuhan Institute of Biological Products Co. Ltd., No.1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430207, China
| | - Sheng-Li Meng
- Wuhan Institute of Biological Products Co. Ltd., No.1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430207, China
| | - Jing Guo
- Wuhan Institute of Biological Products Co. Ltd., No.1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430207, China
| | - Ze-Jun Wang
- Wuhan Institute of Biological Products Co. Ltd., No.1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430207, China
| | - Shuo Shen
- Wuhan Institute of Biological Products Co. Ltd., No.1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430207, China.
| |
Collapse
|
24
|
Flynn JA, Weber T, Cejas PJ, Cox KS, Touch S, Austin LA, Ou Y, Citron MP, Luo B, Gindy ME, Bahl K, Ciaramella G, Espeseth AS, Zhang L. Characterization of humoral and cell-mediated immunity induced by mRNA vaccines expressing influenza hemagglutinin stem and nucleoprotein in mice and nonhuman primates. Vaccine 2022; 40:4412-4423. [PMID: 35680500 DOI: 10.1016/j.vaccine.2022.03.063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 02/24/2022] [Accepted: 03/27/2022] [Indexed: 10/18/2022]
Abstract
In response to immune pressure, influenza viruses evolve, producing drifted variants capable of escaping immune recognition. One strategy for inducing a broad-spectrum immune response capable of recognizing multiple antigenically diverse strains is to target conserved proteins or protein domains. To that end, we assessed the efficacy and immunogenicity of mRNA vaccines encoding either the conserved stem domain of a group 1 hemagglutinin (HA), a group 2 nucleoprotein (NP), or a combination of the two antigens in mice, as well as evaluated immunogenicity in naïve and influenza seropositive nonhuman primates (NHPs). HA stem-immunized animals developed a robust anti-stem antibody binding titer, and serum antibodies recognized antigenically distinct group 1 HA proteins. These antibodies showed little to no neutralizing activity in vitro but were active in an assay measuring induction of antibody-dependent cellular cytotoxicity. HA-directed cell-mediated immunity was weak following HA stem mRNA vaccination; however, robust CD4 and CD8 T cell responses were detected in both mice and NHPs after immunization with mRNA vaccines encoding NP. Both HA stem and NP mRNA vaccines partially protected mice from morbidity following lethal influenza virus challenge, and superior efficacy against two different H1N1 strains was observed when the antigens were combined. In vivo T cell depletion suggested that anti-NP cell-mediated immunity contributed to protection in the mouse model. Taken together, these data show that mRNA vaccines encoding conserved influenza antigens, like HA stem and NP in combination, induce broadly reactive humoral responses as well as cell-mediated immunity in mice and NHPs, providing protection against homologous and heterologous influenza infection in mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yangsi Ou
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - Bin Luo
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | | | | | - Lan Zhang
- Merck & Co., Inc., Kenilworth, NJ, USA.
| |
Collapse
|
25
|
Tharmalingam T, Han X, Wozniak A, Saward L. Polyclonal hyper immunoglobulin: A proven treatment and prophylaxis platform for passive immunization to address existing and emerging diseases. Hum Vaccin Immunother 2022; 18:1886560. [PMID: 34010089 PMCID: PMC9090292 DOI: 10.1080/21645515.2021.1886560] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/26/2021] [Accepted: 02/01/2021] [Indexed: 12/13/2022] Open
Abstract
Passive immunization with polyclonal hyper immunoglobulin (HIG) therapy represents a proven strategy by transferring immunoglobulins to patients to confer immediate protection against a range of pathogens including infectious agents and toxins. Distinct from active immunization, the protection is passive and the immunoglobulins will clear from the system; therefore, administration of an effective dose must be maintained for prophylaxis or treatment until a natural adaptive immune response is mounted or the pathogen/agent is cleared. The current review provides an overview of this technology, key considerations to address different pathogens, and suggested improvements. The review will reflect on key learnings from development of HIGs in the response to public health threats due to Zika, influenza, and severe acute respiratory syndrome coronavirus 2.
Collapse
Affiliation(s)
- Tharmala Tharmalingam
- Therapeutics Business Unit, Emergent BioSolutions Incorporated, Winnipeg, MB, Canada
| | - Xiaobing Han
- Therapeutics Business Unit, Emergent BioSolutions Incorporated, Winnipeg, MB, Canada
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Ashley Wozniak
- Therapeutics Business Unit, Emergent BioSolutions Incorporated, Winnipeg, MB, Canada
| | - Laura Saward
- Therapeutics Business Unit, Emergent BioSolutions Incorporated, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
26
|
He Q, Mao Q, Peng X, He Z, Lu S, Zhang J, Gao F, Bian L, An C, Yu W, Yang F, Zhou Y, Yang Y, Li Y, Yuan Y, Yan X, Yang J, Wu X, Huang W, Li C, Wang J, Liang Z, Xu M. Immunogenicity and protective efficacy of a recombinant protein subunit vaccine and an inactivated vaccine against SARS-CoV-2 variants in non-human primates. Signal Transduct Target Ther 2022; 7:69. [PMID: 35241645 PMCID: PMC8892123 DOI: 10.1038/s41392-022-00926-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/07/2022] [Accepted: 02/13/2022] [Indexed: 02/07/2023] Open
Abstract
Emerging SARS-CoV-2 variants and the gradually decreasing neutralizing antibodies over time post vaccination have led to an increase in incidents of breakthrough infection across the world. To investigate the potential protective effect of the recombinant protein subunit COVID-19 vaccine targeting receptor-binding domain (RBD) (PS-RBD) and whole inactivated virus particle vaccine (IV) against the variant strains, in this study, rhesus macaques were immunized with PS-RBD or IV vaccine, followed by a Beta variant (B.1.351) challenge. Although neutralizing activity against the Beta variant was reduced compared with that against the prototype, the decreased viral load in both upper and lower respiratory tracts, milder pathological changes, and downregulated inflammatory cytokine levels in lung tissues after challenge demonstrated that PS-RBD and IV still provided effective protection against the Beta variant in the macaque model. Furthermore, PS-RBD-induced macaque sera possessed general binding and neutralizing activity to Alpha, Beta, Delta, and Omicron variants in our study, though the neutralizing antibody (NAb) titers declined by varying degrees, demonstrating potential protection of PS-RBD against current circulating variants of concern (VOCs). Interestingly, although the IV vaccine-induced extremely low neutralizing antibody titers against the Beta variant, it still showed reduction for viral load and significantly alleviated pathological change. Other correlates of vaccine-induced protection (CoP) like antibody-dependent cellular cytotoxicity (ADCC) and immune memory were both confirmed to be existing in IV vaccinated group and possibly be involved in the protective mechanism.
Collapse
Affiliation(s)
- Qian He
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China
| | - Qunying Mao
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China
| | - Xiaozhong Peng
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Kunming, 650118, People's Republic of China
| | - Zhanlong He
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Kunming, 650118, People's Republic of China
| | - Shuaiyao Lu
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Kunming, 650118, People's Republic of China
| | - Jialu Zhang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China
| | - Fan Gao
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China
| | - Lianlian Bian
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China
| | - Chaoqiang An
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China
| | - Wenhai Yu
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Kunming, 650118, People's Republic of China
| | - Fengmei Yang
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Kunming, 650118, People's Republic of China
| | - Yanan Zhou
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Kunming, 650118, People's Republic of China
| | - Yun Yang
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Kunming, 650118, People's Republic of China
| | - Yanyan Li
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Kunming, 650118, People's Republic of China
| | - Yadi Yuan
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China
| | - Xujia Yan
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China
| | - Jinghuan Yang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China
| | - Xing Wu
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China
| | - Weijin Huang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China
| | - Changgui Li
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China
| | - Junzhi Wang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China.
| | - Zhenglun Liang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China.
| | - Miao Xu
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control; NHC Key Laboratory of Research on Quality and Standardization of Biotech Products; NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, 102629, People's Republic of China.
| |
Collapse
|
27
|
Dong C, Wang BZ. Engineered Nanoparticulate Vaccines to Combat Recurring and Pandemic Influenza Threats. ADVANCED NANOBIOMED RESEARCH 2022; 2:2100122. [PMID: 35754779 PMCID: PMC9231845 DOI: 10.1002/anbr.202100122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Reoccurring seasonal flu epidemics and occasional pandemics are among the most severe threats to public health. Current seasonal influenza vaccines provide limited protection against drifted circulating strains and no protection against influenza pandemics. Next-generation influenza vaccines, designated as universal influenza vaccines, should be safe, affordable, and elicit long-lasting cross-protective influenza immunity. Nanotechnology plays a critical role in the development of such novel vaccines. Engineered nanoparticles can incorporate multiple advantageous properties into the same nanoparticulate platforms to improve vaccine potency and breadth. These immunological properties include virus-like biomimicry, high antigen-load, controlled antigen release, targeted delivery, and induction of innate signaling pathways. Many nanoparticle influenza vaccines have shown promising results in generating potent and broadly protective immune responses. This review will summarize the necessity and characteristics of next-generation influenza vaccines and the immunological correlates of broad influenza immunity and focus on how cutting-edge nanoparticle technology contributes to such vaccine development. The review will give new insights into the rational design of nanoparticle universal vaccines to combat influenza epidemics and pandemics.
Collapse
Affiliation(s)
- Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia 30303, USA
| |
Collapse
|
28
|
Dong C, Wang Y, Zhu W, Ma Y, Kim J, Wei L, Gonzalez GX, Wang BZ. Polycationic HA/CpG Nanoparticles Induce Cross-Protective Influenza Immunity in Mice. ACS APPLIED MATERIALS & INTERFACES 2022; 14:6331-6342. [PMID: 35084819 PMCID: PMC8832387 DOI: 10.1021/acsami.1c19192] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/20/2021] [Indexed: 05/28/2023]
Abstract
The intranasal (i.n.) route is an ideal vaccination approach for infectious respiratory diseases like influenza. Polycationic polyethylenimine (PEI) could form nanoscale complexes with negatively charged viral glycoproteins. Here we fabricated PEI-hemagglutinin (HA) and PEI-HA/CpG nanoparticles and investigated their immune responses and protective efficacies with an i.n. vaccination regimen in mice. Our results revealed that the nanoparticles significantly enhanced HA immunogenicity, providing heterologous cross-protection. The conserved HA stalk region induced substantial antibodies in the nanoparticle immunization groups. In contrast to the Th2-biased, IgG1-dominant antibody response generated by PEI-HA nanoparticles, PEI-HA/CpG nanoparticles generated more robust and balanced IgG1/IgG2a antibody responses with augmented neutralization activity and Fc-mediated antibody-dependent cellular cytotoxicity (ADCC). PEI-HA/CpG nanoparticles also induced enhanced local and systemic cellular immune responses. These immune responses did not decay over six months of observation postimmunization. PEI and CpG synergized these comprehensive immune responses. Thus, the PEI-HA/CpG nanoparticle is a potential cross-protective influenza vaccine candidate. Polycationic PEI nanoplatforms merit future development into mucosal vaccine systems.
Collapse
|
29
|
Boudreau CM, Burke JS, Shuey KD, Wolf C, Katz J, Tielsch J, Khatry S, LeClerq SC, Englund JA, Chu HY, Alter G. Dissecting Fc signatures of protection in neonates following maternal influenza vaccination in a placebo-controlled trial. Cell Rep 2022; 38:110337. [PMID: 35139373 PMCID: PMC9026287 DOI: 10.1016/j.celrep.2022.110337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/19/2021] [Accepted: 01/13/2022] [Indexed: 11/24/2022] Open
Abstract
Influenza is an important cause of illness and morbidity for infants. Seasonal influenza vaccination during pregnancy aims to provide protection to mothers, but it can also provide immunity to infants. The precise influence of maternal vaccination on immunity in infants and how vaccine-elicited antibodies provide protection in some but not all infants is incompletely understood. We comprehensively profiled the transfer of functional antibodies and defined humoral factors contributing to immunity against influenza in a clinical trial of maternal influenza vaccination. Influenza-specific antibody subclass levels, Fc ɣ receptor (FCGR) binding levels, and antibody-dependent innate immune functions were all profiled in the mothers during pregnancy and at birth, as well as in cord blood. Vaccination increased influenza-specific antibody levels, antibody binding to FCGR, and specific antibody-dependent innate immune functions in both maternal and cord blood, with FCGR binding most enhanced via vaccination. Influenza-specific FCGR binding levels were lower in cord blood of infants who subsequently developed influenza infection. Collectively these data suggest that in addition to increased antibody amounts, the selective transfer of FCGR-binding antibodies contributes to the protective immune response in infants against influenza.
Collapse
Affiliation(s)
- Carolyn M Boudreau
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; PhD Program in Virology, Harvard University, Cambridge, MA, USA
| | - John S Burke
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Kiel D Shuey
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Caitlin Wolf
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Joanne Katz
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - James Tielsch
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Subarna Khatry
- Nepal Nutrition Intervention Project, Sarlahi, Kathmandu, Nepal
| | - Steven C LeClerq
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Nepal Nutrition Intervention Project, Sarlahi, Kathmandu, Nepal
| | - Janet A Englund
- Department of Pediatrics, Seattle Children's Research Institute and University of Washington, Seattle, WA, USA.
| | - Helen Y Chu
- Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
30
|
Caradonna TM, Schmidt AG. Protein engineering strategies for rational immunogen design. NPJ Vaccines 2021; 6:154. [PMID: 34921149 PMCID: PMC8683408 DOI: 10.1038/s41541-021-00417-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022] Open
Abstract
Antibody immunodominance refers to the preferential and asymmetric elicitation of antibodies against specific epitopes on a complex protein antigen. Traditional vaccination approaches for rapidly evolving pathogens have had limited success in part because of this phenomenon, as elicited antibodies preferentially target highly variable regions of antigens, and thus do not confer long lasting protection. While antibodies targeting functionally conserved epitopes have the potential to be broadly protective, they often make up a minority of the overall repertoire. Here, we discuss recent protein engineering strategies used to favorably alter patterns of immunodominance, and selectively focus antibody responses toward broadly protective epitopes in the pursuit of next-generation vaccines for rapidly evolving pathogens.
Collapse
Affiliation(s)
| | - Aaron G Schmidt
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
31
|
Lassaunière R, Tiemessen CT. FcγR Genetic Variation and HIV-1 Vaccine Efficacy: Context And Considerations. Front Immunol 2021; 12:788203. [PMID: 34975881 PMCID: PMC8714752 DOI: 10.3389/fimmu.2021.788203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/29/2021] [Indexed: 12/02/2022] Open
Abstract
Receptors for the crystallisable fragment (Fc) of immunoglobulin (Ig) G, Fcγ receptors (FcγRs), link the humoral and cellular arms of the immune response, providing a diverse armamentarium of antimicrobial effector functions. Findings from HIV-1 vaccine efficacy trials highlight the need for further study of Fc-FcR interactions in understanding what may constitute vaccine-induced protective immunity. These include host genetic correlates identified within the low affinity Fcγ-receptor locus in three HIV-1 efficacy trials – VAX004, RV144, and HVTN 505. This perspective summarizes our present knowledge of FcγR genetics in the context of findings from HIV-1 efficacy trials, and draws on genetic variation described in other contexts, such as mother-to-child HIV-1 transmission and HIV-1 disease progression, to explore the potential contribution of FcγR variability in modulating different HIV-1 vaccine efficacy outcomes. Appreciating the complexity and the importance of the collective contribution of variation within the FCGR gene locus is important for understanding the role of FcγRs in protection against HIV-1 acquisition.
Collapse
Affiliation(s)
- Ria Lassaunière
- Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
- *Correspondence: Caroline T. Tiemessen, ; Ria Lassaunière,
| | - Caroline T. Tiemessen
- Centre for HIV and STI’s, National Institute for Communicable Diseases, Johannesburg, South Africa
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- *Correspondence: Caroline T. Tiemessen, ; Ria Lassaunière,
| |
Collapse
|
32
|
Yu Y, Wang M, Zhang X, Li S, Lu Q, Zeng H, Hou H, Li H, Zhang M, Jiang F, Wu J, Ding R, Zhou Z, Liu M, Si W, Zhu T, Li H, Ma J, Gu Y, She G, Li X, Zhang Y, Peng K, Huang W, Liu W, Wang Y. Antibody-dependent cellular cytotoxicity response to SARS-CoV-2 in COVID-19 patients. Signal Transduct Target Ther 2021; 6:346. [PMID: 34561414 PMCID: PMC8463587 DOI: 10.1038/s41392-021-00759-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/05/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023] Open
Abstract
Antibody-dependent cellular cytotoxicity (ADCC) responses to viral infection are a form of antibody regulated immune responses mediated through the Fc fragment. Whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) triggered ADCC responses contributes to COVID-19 disease development is currently not well understood. To understand the potential correlation between ADCC responses and COVID-19 disease development, we analyzed the ADCC activity and neutralizing antibody response in 255 individuals ranging from asymptomatic to fatal infections over 1 year post disease. ADCC was elicited by 10 days post-infection, peaked by 11-20 days, and remained detectable until 400 days post-infection. In general, patients with severe disease had higher ADCC activities. Notably, patients who had severe disease and recovered had higher ADCC activities than patients who had severe disease and deceased. Importantly, ADCC activities were mediated by a diversity of epitopes in SARS-COV-2-infected mice and induced to comparable levels against SARS-CoV-2 variants of concern (VOCs) (B.1.1.7, B.1.351, and P.1) as that against the D614G mutant in human patients and vaccinated mice. Our study indicates anti-SARS-CoV-2 ADCC as a major trait of COVID-19 patients with various conditions, which can be applied to estimate the extra-neutralization level against COVID-19, especially lethal COVID-19.
Collapse
Affiliation(s)
- Yuanling Yu
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Meiyu Wang
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China ,grid.506261.60000 0001 0706 7839Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaoai Zhang
- grid.410740.60000 0004 1803 4911State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Shufen Li
- grid.9227.e0000000119573309State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei China
| | - Qingbin Lu
- grid.11135.370000 0001 2256 9319Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, China
| | - Haolong Zeng
- grid.33199.310000 0004 0368 7223Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyan Hou
- grid.33199.310000 0004 0368 7223Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Li
- grid.410740.60000 0004 1803 4911State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Mengyi Zhang
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Fei Jiang
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Jiajing Wu
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Ruxia Ding
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Zehua Zhou
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Min Liu
- grid.274690.eSinovac Biotech Co., Ltd, Beijing, China
| | - Weixue Si
- Cansino Biotech Incorporation, Tianjin, China
| | - Tao Zhu
- Cansino Biotech Incorporation, Tianjin, China
| | - Hangwen Li
- Stemirna Therapeutics, Ltd, Shanghai, China
| | - Jie Ma
- Stemirna Therapeutics, Ltd, Shanghai, China
| | | | - Guangbiao She
- Anhui Zhifeilongcom Biopharmaceutical Co., Ltd, Hefei, China
| | - Xiaokun Li
- grid.410740.60000 0004 1803 4911State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yulan Zhang
- grid.9227.e0000000119573309State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei China
| | - Ke Peng
- grid.9227.e0000000119573309State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Weijin Huang
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Wei Liu
- grid.410740.60000 0004 1803 4911State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Youchun Wang
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China ,grid.506261.60000 0001 0706 7839Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
33
|
Huseby Kelcher AM, Baehr CA, Hamid FA, Hart GT, Pravetoni M. Contribution of Antibody-Mediated Effector Functions to the Mechanism of Efficacy of Vaccines for Opioid Use Disorders. THE JOURNAL OF IMMUNOLOGY 2021; 207:860-867. [PMID: 34281999 DOI: 10.4049/jimmunol.2100204] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/28/2021] [Indexed: 12/28/2022]
Abstract
Vaccines and mAbs offer promising strategies to treat substance use disorders (SUDs) and prevent overdose. Despite vaccines and mAbs against SUDs demonstrating proof of efficacy, selectivity, and safety in animal models, it is unknown whether the mechanism of action of these immunotherapeutics relies exclusively on the formation of Ab/drug complexes, or also involves Ab-mediated effector functions. Hence, this study tested whether the efficacy of active and passive immunization against drugs of abuse requires phagocytosis, the intact Fc portion of the anti-drug Ab, FcγRs, or the neonatal FcR (FcRn). The efficacy of a lead vaccine against oxycodone was not diminished in mice after depletion of macrophages or granulocytes. Anti-oxycodone F(ab')2 fragments resulted in lower serum levels of F(ab')2 compared with intact mAbs, and F(ab')2s were not as effective as the parent mAbs in reducing distribution of oxycodone to the brain. The efficacy of vaccines and mAbs against oxycodone was preserved in either FcγIII or FcγI-IV ablated mice, suggesting that FcγRs are not required for Ab efficacy. Finally, both active and passive immunization against oxycodone in FcRn-/- mice yielded reduced efficacy compared with wild-type control mice. These data identified a role for FcRn, but not for phagocytosis or Fc-dependent effector functions, in mediating the efficacy of vaccines and mAbs against SUD. This study supports rational design of vaccines and mAbs engineered for maximal neutralization activity and optimal FcRn binding.
Collapse
Affiliation(s)
| | - Carly A Baehr
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN
| | - Fatima A Hamid
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN
| | - Geoffrey T Hart
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN; and.,Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
| | - Marco Pravetoni
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN; .,Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
| |
Collapse
|
34
|
Suscovich TJ, Fallon JK, Das J, Demas AR, Crain J, Linde CH, Michell A, Natarajan H, Arevalo C, Broge T, Linnekin T, Kulkarni V, Lu R, Slein MD, Luedemann C, Marquette M, March S, Weiner J, Gregory S, Coccia M, Flores-Garcia Y, Zavala F, Ackerman ME, Bergmann-Leitner E, Hendriks J, Sadoff J, Dutta S, Bhatia SN, Lauffenburger DA, Jongert E, Wille-Reece U, Alter G. Mapping functional humoral correlates of protection against malaria challenge following RTS,S/AS01 vaccination. Sci Transl Med 2021; 12:12/553/eabb4757. [PMID: 32718991 DOI: 10.1126/scitranslmed.abb4757] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022]
Abstract
Vaccine development has the potential to be accelerated by coupling tools such as systems immunology analyses and controlled human infection models to define the protective efficacy of prospective immunogens without expensive and slow phase 2b/3 vaccine studies. Among human challenge models, controlled human malaria infection trials have long been used to evaluate candidate vaccines, and RTS,S/AS01 is the most advanced malaria vaccine candidate, reproducibly demonstrating 40 to 80% protection in human challenge studies in malaria-naïve individuals. Although antibodies are critical for protection after RTS,S/AS01 vaccination, antibody concentrations are inconsistently associated with protection across studies, and the precise mechanism(s) by which vaccine-induced antibodies provide protection remains enigmatic. Using a comprehensive systems serological profiling platform, the humoral correlates of protection against malaria were identified and validated across multiple challenge studies. Rather than antibody concentration, qualitative functional humoral features robustly predicted protection from infection across vaccine regimens. Despite the functional diversity of vaccine-induced immune responses across additional RTS,S/AS01 vaccine studies, the same antibody features, antibody-mediated phagocytosis and engagement of Fc gamma receptor 3A (FCGR3A), were able to predict protection across two additional human challenge studies. Functional validation using monoclonal antibodies confirmed the protective role of Fc-mediated antibody functions in restricting parasite infection both in vitro and in vivo, suggesting that these correlates may mechanistically contribute to parasite restriction and can be used to guide the rational design of an improved vaccine against malaria.
Collapse
Affiliation(s)
- Todd J Suscovich
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | | | - Jishnu Das
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Allison R Demas
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jonathan Crain
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Caitlyn H Linde
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Ashlin Michell
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Harini Natarajan
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Claudia Arevalo
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Thomas Broge
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Thomas Linnekin
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Viraj Kulkarni
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Richard Lu
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Matthew D Slein
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | | | - Meghan Marquette
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sandra March
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joshua Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Scott Gregory
- PATH's Malaria Vaccine Initiative, Washington, DC 20001, USA
| | | | - Yevel Flores-Garcia
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Fidel Zavala
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Elke Bergmann-Leitner
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Jenny Hendriks
- Janssen Vaccines & Prevention B.V., 2333CN Leiden, Netherlands
| | - Jerald Sadoff
- Janssen Vaccines & Prevention B.V., 2333CN Leiden, Netherlands
| | - Sheetij Dutta
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA.,Broad Institute, Cambridge, MA 02139, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Douglas A Lauffenburger
- Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | - Galit Alter
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
35
|
Ng KW, Faulkner N, Wrobel AG, Gamblin SJ, Kassiotis G. Heterologous humoral immunity to human and zoonotic coronaviruses: Aiming for the achilles heel. Semin Immunol 2021; 55:101507. [PMID: 34716096 PMCID: PMC8542444 DOI: 10.1016/j.smim.2021.101507] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 02/04/2023]
Abstract
Coronaviruses are evolutionarily successful RNA viruses, common to multiple avian, amphibian and mammalian hosts. Despite their ubiquity and potential impact, knowledge of host immunity to coronaviruses remains incomplete, partly owing to the lack of overt pathogenicity of endemic human coronaviruses (HCoVs), which typically cause common colds. However, the need for deeper understanding became pressing with the zoonotic introduction of three novel coronaviruses in the past two decades, causing severe acute respiratory syndromes in humans, and the unfolding pandemic of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This renewed interest not only triggered the discovery of two of the four HCoVs, but also uncovered substantial cellular and humoral cross-reactivity with shared or related coronaviral antigens. Here, we review the evidence for cross-reactive B cell memory elicited by HCoVs and its potential impact on the puzzlingly variable outcome of SARS-CoV-2 infection. The available data indicate targeting of highly conserved regions primarily in the S2 subunits of the spike glycoproteins of HCoVs and SARS-CoV-2 by cross-reactive B cells and antibodies. Rare monoclonal antibodies reactive with conserved S2 epitopes and with potent virus neutralising activity have been cloned, underscoring the potential functional relevance of cross-reactivity. We discuss B cell and antibody cross-reactivity in the broader context of heterologous humoral immunity to coronaviruses, as well as the limits of protective immune memory against homologous re-infection. Given the bidirectional nature of cross-reactivity, the unprecedented current vaccination campaign against SARS-CoV-2 is expected to impact HCoVs, as well as future zoonotic coronaviruses attempting to cross the species barrier. However, emerging SARS-CoV-2 variants with resistance to neutralisation by vaccine-induced antibodies highlight a need for targeting more constrained, less mutable parts of the spike. The delineation of such cross-reactive areas, which humoral immunity can be trained to attack, may offer the key to permanently shifting the balance of our interaction with current and future coronaviruses in our favour.
Collapse
Affiliation(s)
- Kevin W Ng
- Retroviral Immunology Laboratory, London, NW1 1AT, UK
| | - Nikhil Faulkner
- Retroviral Immunology Laboratory, London, NW1 1AT, UK; National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Antoni G Wrobel
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Steve J Gamblin
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - George Kassiotis
- Retroviral Immunology Laboratory, London, NW1 1AT, UK; Department of Infectious Disease, St Mary's Hospital, Imperial College London, London W2 1PG, UK.
| |
Collapse
|
36
|
Sanchez Vargas LA, Adam A, Masterson M, Smith M, Lyski ZL, Dowd KA, Pierson TC, Messer WB, Currier JR, Mathew A. Non-structural protein 1-specific antibodies directed against Zika virus in humans mediate antibody-dependent cellular cytotoxicity. Immunology 2021; 164:386-397. [PMID: 34056709 PMCID: PMC8442231 DOI: 10.1111/imm.13380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/18/2021] [Accepted: 05/23/2021] [Indexed: 12/14/2022] Open
Abstract
There is growing interest in understanding antibody (Ab) function beyond neutralization. The non-structural protein 1 (NS1) of Zika virus (ZIKV) is an attractive candidate for an effective vaccine as Abs against NS1, unlike the envelope or premembrane, do not carry the risk of mediating antibody-dependent enhancement. Our aim was to evaluate whether ZIKV NS1 Abs elicited following natural infection in humans can mediate antibody-dependent cellular cytotoxicity (ADCC). We evaluated the isotype specificity of ZIKV-specific Abs in immune sera and supernatants from stimulated immune PBMC and found that Abs against ZIKV NS1 and virus-like particles were predominantly of the IgG1 isotype. Using a recently developed FluoroSpot assay, we found robust frequencies of NS1-specific Ab-secreting cells in PBMC of individuals who were naturally infected with ZIKV. We developed assays to measure both natural killer cell activation by flow cytometry and target cell lysis of ZIKV NS1-expressing cells using an image cytometry assay in the presence of ZIKV NS1 Abs. Our data indicate efficient opsonization of ZIKV NS1-expressing CEM-NKR cell lines using ZIKV-immune but not ZIKV-naïve sera, a prerequisite of ADCC. Furthermore, sera from immune donors were able to induce both NK cell degranulation and lysis of ZIKV NS1 CEM-NKR cells in vitro. Our data suggest that ADCC is a possible mechanism for ZIKV NS1 Abs to eliminate virally infected target cells.
Collapse
Affiliation(s)
- Luis A Sanchez Vargas
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, USA
| | - Awadalkareem Adam
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, USA
| | - Mary Masterson
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Madison Smith
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, USA
| | - Zoe L Lyski
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | | | | | - William B Messer
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA.,Division of Infectious Diseases, Department of Medicine, Oregon Health & Science University, Portland, OR, USA.,OHSU-PSU School of Public Health, Program in Epidemiology, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey R Currier
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Anuja Mathew
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, USA
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Despite its crucial role in protection against viral infections, mucosal immunity has been largely understudied in the context of coronavirus disease 2019 (COVID-19). This review outlines the current evidence about the role of mucosal immune responses in the clearance of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, as well as potential mucosal mechanisms of protection against (re-)infection. RECENT FINDINGS The angiotensin-converting enzyme 2 cellular entry receptor for SARS-CoV-2 is most highly expressed in the upper respiratory tract and most SARS-CoV-2 shedding occurs from the upper respiratory tract. Viral shedding peaks early during infection around the onset of symptoms, before dropping rapidly in most individuals within 7 days of symptom onset, suggesting mucosal inhibition of viral infection. Serum and mucosal immunoglobulin G and immunoglobulin M responses were found to be strongly correlated in infected patients, whereas correlations were much weaker for immunoglobulin A (IgA). Mucosal IgA responses have been detected in infected cases in the absence of serum antibody responses, with mucosal antibody levels correlating strongly with virus neutralization. Bulk and single-cell RNA sequencing analysis of nasopharyngeal swabs and bronchoalveolar lavage samples of COVID-19 patients revealed the induction of mucosal chemokine and cytokine genes, complement pathways, Janus Kinase/Signal Transducer and Activator of Transcription signaling and cytotoxic T cells. SUMMARY Although most clinical studies focus on antibodies and cellular immunity in peripheral blood, mucosal immune responses in the respiratory tract play a key role in the early restriction of viral replication and the clearance of SARS-CoV-2. Identification of mucosal biomarkers associated with viral clearance will allow monitoring of infection-induced immunity. Further studies are needed to understand how the systemic immunological endpoints measured in vaccination studies translate to mucosal protection against SARS-CoV-2 infection.
Collapse
|
38
|
Tung Yep A, Takeuchi Y, Engelhardt OG, Hufton SE. Broad Reactivity Single Domain Antibodies against Influenza Virus and Their Applications to Vaccine Potency Testing and Immunotherapy. Biomolecules 2021; 11:biom11030407. [PMID: 33802072 PMCID: PMC8001348 DOI: 10.3390/biom11030407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/05/2021] [Accepted: 03/06/2021] [Indexed: 12/11/2022] Open
Abstract
The antigenic variability of influenza presents many challenges to the development of vaccines and immunotherapeutics. However, it is apparent that there are epitopes on the virus that have evolved to remain largely constant due to their functional importance. These more conserved regions are often hidden and difficult to access by the human immune system but recent efforts have shown that these may be the Achilles heel of the virus through development and delivery of appropriate biological drugs. Amongst these, single domain antibodies (sdAbs) are equipped to target these vulnerabilities of the influenza virus due to their preference for concave epitopes on protein surfaces, their small size, flexible reformatting and high stability. Single domain antibodies are well placed to provide a new generation of robust analytical reagents and therapeutics to support the constant efforts to keep influenza in check.
Collapse
Affiliation(s)
- Andrew Tung Yep
- Biotherapeutics Division, National Institute for Biological Standards and Control (NIBSC), Potters Bar, Hertfordshire EN6 3QG, UK;
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK;
| | - Yasu Takeuchi
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK;
- Advanced Therapies Division, NIBSC, Potters Bar, Hertfordshire EN6 3QG, UK
| | | | - Simon E. Hufton
- Biotherapeutics Division, National Institute for Biological Standards and Control (NIBSC), Potters Bar, Hertfordshire EN6 3QG, UK;
- Correspondence:
| |
Collapse
|
39
|
Klein SL, Pekosz A, Park HS, Ursin RL, Shapiro JR, Benner SE, Littlefield K, Kumar S, Naik HM, Betenbaugh MJ, Shrestha R, Wu AA, Hughes RM, Burgess I, Caturegli P, Laeyendecker O, Quinn TC, Sullivan D, Shoham S, Redd AD, Bloch EM, Casadevall A, Tobian AA. Sex, age, and hospitalization drive antibody responses in a COVID-19 convalescent plasma donor population. J Clin Invest 2020; 130:6141-6150. [PMID: 32764200 PMCID: PMC7598041 DOI: 10.1172/jci142004] [Citation(s) in RCA: 364] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
Convalescent plasma is a leading treatment for coronavirus disease 2019 (COVID-19), but there is a paucity of data identifying its therapeutic efficacy. Among 126 potential convalescent plasma donors, the humoral immune response was evaluated using a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus neutralization assay with Vero-E6-TMPRSS2 cells; a commercial IgG and IgA ELISA to detect the spike (S) protein S1 domain (EUROIMMUN); IgA, IgG, and IgM indirect ELISAs to detect the full-length S protein or S receptor-binding domain (S-RBD); and an IgG avidity assay. We used multiple linear regression and predictive models to assess the correlations between antibody responses and demographic and clinical characteristics. IgG titers were greater than either IgM or IgA titers for S1, full-length S, and S-RBD in the overall population. Of the 126 plasma samples, 101 (80%) had detectable neutralizing antibody (nAb) titers. Using nAb titers as the reference, the IgG ELISAs confirmed 95%-98% of the nAb-positive samples, but 20%-32% of the nAb-negative samples were still IgG ELISA positive. Male sex, older age, and hospitalization for COVID-19 were associated with increased antibody responses across the serological assays. There was substantial heterogeneity in the antibody response among potential convalescent plasma donors, but sex, age, and hospitalization emerged as factors that can be used to identify individuals with a high likelihood of having strong antiviral antibody responses.
Collapse
Affiliation(s)
- Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology
- Department of Biochemistry and Molecular Biology
- Department of International Health, and
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Han-Sol Park
- W. Harry Feinstone Department of Molecular Microbiology and Immunology
| | | | | | - Sarah E. Benner
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | - Swetha Kumar
- Advanced Mammalian Biomanufacturing Innovation Center, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Harnish Mukesh Naik
- Advanced Mammalian Biomanufacturing Innovation Center, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michael J. Betenbaugh
- Advanced Mammalian Biomanufacturing Innovation Center, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ruchee Shrestha
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Annie A. Wu
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Robert M. Hughes
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Imani Burgess
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Patricio Caturegli
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Oliver Laeyendecker
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Thomas C. Quinn
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - David Sullivan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Andrew D. Redd
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Evan M. Bloch
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Arturo Casadevall
- W. Harry Feinstone Department of Molecular Microbiology and Immunology
| | - Aaron A.R. Tobian
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
40
|
Vangelista L, Secchi M. Prepare for the Future: Dissecting the Spike to Seek Broadly Neutralizing Antibodies and Universal Vaccine for Pandemic Coronaviruses. Front Mol Biosci 2020; 7:226. [PMID: 33033717 PMCID: PMC7490329 DOI: 10.3389/fmolb.2020.00226] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/11/2020] [Indexed: 01/07/2023] Open
Abstract
Learning from the lengthy fight against HIV-1, influenza, and Ebola virus infection, broadly neutralizing antibodies (bnAbs), directed at conserved regions of surface proteins crucial to virus entry (Env, hemagglutinin, and GP, respectively), are an essential resource for passive as well as active immunization. Rare in their emergence and antigen recognition mode, bnAbs are active toward a large set of different viral strains. Isolation, characterization and production of bnAbs lead to their possible use in passive immunotherapy and form the basis for an educated effort in the development of vaccines for universal coverage. SARS-CoV-2-specific antibodies targeting the spike receptor binding domain (RBD) may lead to antibody dependent enhancement (ADE) of infection, possibly hampering the field of vaccine development. This perspective points to the identification of conserved regions in the spike of SARS-CoV-2, SARS-CoV, and MERS-CoV through investigation, dissection and recombinant production of isolated moieties. These spike moieties should be capable of independent folding and allow the detection as well as the elicitation of bnAbs, thus setting the basis for an effective passive immunotherapy and the development of a universal vaccine against human epidemic coronaviruses (HCoVs). SARS, MERS and, most of all, COVID-19 demonstrate that humanity is the target of HCoV, preparedness for future hits is thus no longer an option.
Collapse
Affiliation(s)
- Luca Vangelista
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Nur-Sultan, Kazakhstan
| | | |
Collapse
|
41
|
Hutchinson NT, Steelman A, Woods JA. Behavioral strategies to prevent and mitigate COVID-19 infection. SPORTS MEDICINE AND HEALTH SCIENCE 2020; 2:115-125. [PMID: 34189481 PMCID: PMC7481129 DOI: 10.1016/j.smhs.2020.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022] Open
Abstract
The single stranded RNA virus SARS-CoV-2 has caused a massive addition to the already leading global cause of mortality, viral respiratory tract infections. Characterized by and associated with early and deleteriously enhanced production of pro-inflammatory cytokines by respiratory epithelial cells, severe COVID-19 illness has the potential to inflict acute respiratory distress syndrome and even death. Due to the fast spreading nature of COVID-19 and the current lack of a vaccine or specific pharmaceutical treatments, understanding of viral pathogenesis, behavioral prophylaxis, and mitigation tactics are of great public health concern. This review article outlines the immune response to viral pathogens, and due to the novelty of COVID-19 and the large body of evidence suggesting the respiratory and immune benefits from regular moderate intensity exercise, provides observational and mechanistic evidence from research on other viral infections that suggests strategically planned exercise regimens may help reduce susceptibility to infection, while also mitigating severe immune responses to infection commonly associated with poor COVID-19 prognosis. We propose that regular moderate intensity exercise should be considered as part of a combinatorial approach including widespread hygiene initiatives, properly planned and well-executed social distancing policies, and use of efficacious facial coverings like N95 respirators. Studies discerning COVID-19 pathogenesis mechanisms, transfer dynamics, and individual responses to pharmaceutical and adjunct treatments are needed to reduce viral transmission and bring an end to the COVID-19 pandemic.
Collapse
Affiliation(s)
- Noah T. Hutchinson
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Andrew Steelman
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Champaign, IL, USA
- Carle R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Jeffrey A. Woods
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| |
Collapse
|
42
|
Klein S, Pekosz A, Park HS, Ursin R, Shapiro J, Benner S, Littlefield K, Kumar S, Naik HM, Betenbaugh M, Shrestha R, Wu A, Hughes R, Burgess I, Caturegli P, Laeyendecker O, Quinn T, Sullivan D, Shoham S, Redd A, Bloch E, Casadevall A, Tobian A. Sex, age, and hospitalization drive antibody responses in a COVID-19 convalescent plasma donor population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.06.26.20139063. [PMID: 32607519 PMCID: PMC7325184 DOI: 10.1101/2020.06.26.20139063] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Convalescent plasma is currently one of the leading treatments for COVID-19, but there is a paucity of data identifying therapeutic efficacy. A comprehensive analysis of the antibody responses in potential plasma donors and an understanding of the clinical and demographic factors that drive variant antibody responses is needed. Among 126 potential convalescent plasma donors, the humoral immune response was evaluated by a SARS-CoV-2 virus neutralization assay using Vero-E6-TMPRSS2 cells, commercial IgG and IgA ELISA to Spike (S) protein S1 domain (Euroimmun), IgA, IgG and IgM indirect ELISAs to the full-length S or S-receptor binding domain (S-RBD), and an IgG avidity assay. Multiple linear regression and predictive models were utilized to assess the correlations between antibody responses with demographic and clinical characteristics. IgG titers were greater than either IgM or IgA for S1, full length S, and S-RBD in the overall population. Of the 126 plasma samples, 101 (80%) had detectable neutralizing titers. Using neutralization titer as the reference, the sensitivity of the IgG ELISAs ranged between 95-98%, but specificity was only 20-32%. Male sex, older age, and hospitalization with COVID-19 were all consistently associated with increased antibody responses across the serological assays. Neutralizing antibody titers were reduced over time in contrast to overall antibody responses. There was substantial heterogeneity in the antibody response among potential convalescent plasma donors, but sex, age and hospitalization emerged as factors that can be used to identify individuals with a high likelihood of having strong antiviral antibody levels.
Collapse
|
43
|
Murine Cross-Reactive Nonneutralizing Polyclonal IgG1 Antibodies Induced by Influenza Vaccine Inhibit the Cross-Protective Effect of IgG2 against Heterologous Virus in Mice. J Virol 2020; 94:JVI.00323-20. [PMID: 32269125 DOI: 10.1128/jvi.00323-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/29/2020] [Indexed: 12/13/2022] Open
Abstract
Annual vaccination against influenza viruses is the most reliable and efficient way to prevent and control annual epidemics and protect from severe influenza disease. However, current split influenza vaccines are generally not effective against antigenically mismatched (heterologous) strains. To broaden the protective spectrum of influenza vaccines, adjuvants that can induce cross-reactive antibodies with cross-protection via Fc-mediated effector functions are urgently sought. Although IgG2 antibodies are generally more efficient than IgG1 antibodies in Fc-mediated effector functions, it is not yet clear which IgG isotypes show superior cross-protection against heterologous strains. It also remains unclear whether these IgG isotypes interfere with each other's protective effects. Here, we found that influenza split vaccine adjuvanted with aluminum salts, which predominantly induce cross-reactive IgG1, did not confer cross-protection against heterologous virus challenge in mice. In contrast, split vaccine adjuvanted with CpG oligodeoxynucleotides, which predominantly induce cross-reactive IgG2, showed cross-protection through the interaction of cross-reactive nonneutralizing IgG2 and alveolar macrophages, indicating the importance of cross-reactive nonneutralizing IgG2 for cross-protection. Furthermore, by using serum samples from immunized mice and isolated polyclonal antibodies, we show that vaccine-induced cross-reactive nonneutralizing IgG1 suppress the cross-protective effects of IgG2 by competitively inhibiting the binding of IgG2 to virus. Thus, we demonstrate the new concept that cross-reactive IgG1 may interfere with the potential for cross-protection of influenza vaccine. We propose that adjuvants that selectively induce virus-specific IgG2 in mice, such as CpG oligodeoxynucleotides, are optimal for heterologous protection.IMPORTANCE Current influenza vaccines are generally effective against highly similar virus strains by inducing neutralizing antibodies. However, these antibodies fail to neutralize antigenically mismatched (heterologous) strains and therefore provide limited protection against them. Efforts are being made to develop vaccines with cross-protective ability that would protect broadly against heterologous strains, because the mismatch between predicted and epidemic strains cannot always be avoided, resulting in low vaccine efficacy. Here, we show that nonneutralizing IgG2 antibodies induced by an optimal adjuvant play a crucial role in cross-protection against heterologous virus challenge in mice. Furthermore, nonneutralizing polyclonal IgG1 suppressed the cross-protective effects of nonneutralizing polyclonal IgG2 by competitively blocking the binding of IgG2 to its antigen. These data shed new light on the importance of IgG isotypes and the selection of appropriate adjuvants for the development of universal influenza vaccines. Furthermore, our findings are applicable to the rational design of vaccines against other pathogens.
Collapse
|
44
|
Nguyen-Robertson C, Haque A, Mintern J, La Flamme AC. COVID-19: searching for clues among other respiratory viruses. Immunol Cell Biol 2020; 98:247-250. [PMID: 32319148 DOI: 10.1111/imcb.12336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 01/06/2023]
Affiliation(s)
- Catriona Nguyen-Robertson
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Ashraful Haque
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Justine Mintern
- Department of Biochemistry and Molecular Biology, The University of Melbourne, VIC, Melbourne, Australia
| | - Anne C La Flamme
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
45
|
Chung AW. Multifaceted roles of antibody Fc effector functions: from protection to pathology. Immunol Cell Biol 2020; 98:251-252. [PMID: 32274825 DOI: 10.1111/imcb.12330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 03/16/2020] [Indexed: 12/01/2022]
Affiliation(s)
- Amy W Chung
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| |
Collapse
|
46
|
Madsen MB, Bergsten H, Norrby-Teglund A. Treatment of Necrotizing Soft Tissue Infections: IVIG. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1294:105-125. [DOI: 10.1007/978-3-030-57616-5_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|