1
|
Kardava L, Lim J, Buckner CM, Lopes de Assis F, Zhang X, Wang W, Melnyk ML, El Merhebi O, Trihemasava K, Teng IT, Carroll R, Jethmalani Y, Castro M, Lin BC, Praiss LH, Seamon CA, Kwong PD, Koup RA, Serebryannyy L, Nickle DC, Chun TW, Moir S. Phenotypic heterogeneity defines B cell responses to repeated SARS-CoV-2 exposures through vaccination and infection. Cell Rep 2025; 44:115557. [PMID: 40222009 PMCID: PMC12080740 DOI: 10.1016/j.celrep.2025.115557] [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: 09/23/2024] [Revised: 01/29/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Long-lived humoral memory is key to durable immunity against pathogens yet remains challenging to define due to heterogeneity among antigen-reactive B cells. We addressed this gap through longitudinal sampling over the course of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccinations with or without breakthrough infection. High-dimensional phenotypic profiling performed on ∼72 million B cells showed that receptor-binding domain (RBD) reactivity was associated with five distinct immunoglobulin G (IgG) B cell populations. Two expressed the activation marker CD71, both correlated with neutralizing antibodies, yet the one lacking the memory marker CD27 was induced by vaccination and blunted by infection. Two were resting memory populations; one lacking CD73 arose early and contributed to cross-reactivity; the other, expressing CD73, arose later and correlated with neutralizing antibodies. The fifth, a rare germinal center-like population, contributed to recall responses and was highly cross reactive. Overall, robust and distinct responses to booster vaccination overcame the superiority of hybrid immunity provided by breakthrough infection.
Collapse
Affiliation(s)
- Lela Kardava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - James Lim
- Monoceros Biosystems, San Diego, CA 29130, USA
| | - Clarisa M Buckner
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Felipe Lopes de Assis
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xiaozhen Zhang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Wei Wang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Mattie L Melnyk
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Omar El Merhebi
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Krittin Trihemasava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - I-Ting Teng
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Robin Carroll
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - Mike Castro
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Bob C Lin
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Lauren H Praiss
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Catherine A Seamon
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA
| | - Peter D Kwong
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA; Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Richard A Koup
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - David C Nickle
- Monoceros Biosystems, San Diego, CA 29130, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
2
|
Chu Q, Li K, He Q, Ren L, Wang J, Wang S, Liu X, Liu Y, He J, Li D, Shao Y. Efficient boosting of Omicron-reactive memory B cells after breakthrough infection protects from repeated exposure. iScience 2025; 28:112278. [PMID: 40264792 PMCID: PMC12013488 DOI: 10.1016/j.isci.2025.112278] [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: 01/02/2025] [Revised: 02/17/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Exploring the impact of persistent mutations in SARS-CoV-2 variants and reduced immunity on breakthrough infections (BTIs) is crucial, particularly in understanding how antigen-specific memory B cells (MBCs) respond to new variants. We followed 107 participants who received the ancestral inactivated vaccine and experienced one or two Omicron BTIs over six months. Using flow cytometry, SARS-CoV-2 antigen probes, single-cell RNA sequencing, and B cell receptor (BCR) profiling, we assessed MBCs and immune diversity. Our findings revealed that although neutralizing antibody levels decreased over time, the number of specific MBCs remained stable and matured progressively. Notably, pre-existing Omicron-specific MBCs played a key role in preventing secondary Omicron infections. Differential gene analysis showed enrichment in antigen processing and immune regulation pathways, while clonal lineage analysis revealed more B cell expansion and V(D)J gene-specific rearrangements in high neutralization samples. These results emphasize MBCs' critical role in long-term immunity and inform future vaccination strategies.
Collapse
Affiliation(s)
- Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Kang Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Qianxin He
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Li Ren
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Jiguo Wang
- Toroivd Technology Company Limited, Shanghai 200439, China
| | - Shuo Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Xiaojing Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ying Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Jiangshan He
- College of Life Sciences, Beijing Normal University, 19 Xinjiekouwai Avenue, Beijing 100875, China
| | - Dan Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yiming Shao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Changping Laboratory, Beijing 102299, China
| |
Collapse
|
3
|
Wang X, Kong H, Chu B, Yang Q, Lin C, Liu R, Chen C, Gao Y, Wang G, Wang D, Qin C, Ye X, Yu L, Xu X, Jin J, Sun R, Chen H, Wu X, Zhang Z. Identification of a broad-inhibition influenza neuraminidase antibody from pre-existing memory B cells. Cell Host Microbe 2025; 33:151-166.e8. [PMID: 39740671 DOI: 10.1016/j.chom.2024.12.004] [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: 08/02/2024] [Revised: 09/27/2024] [Accepted: 12/03/2024] [Indexed: 01/02/2025]
Abstract
Identifying broadly reactive B precursor cells and conserved epitopes is crucial for developing a universal flu vaccine. In this study, using influenza neuraminidase (NA) mutant probes, we find that human pre-existing NA-specific memory B cells (MBCs) account for ∼0.25% of total MBCs, which are heterogeneous and dominated by class-unswitched MBCs. In addition, we identify three NA broad-inhibition monoclonal antibodies (mAbs) (BImAbs) that block the activity of NA derived from different influenza strains, including the recent cow H5N1. The cryoelectron microscopy (cryo-EM) structure shows that the BImAb targets the conserved NA enzymatic pocket and a separate epitope in the neighboring NA monomer. Furthermore, the NA BImAbs protect mice from the lethal challenge of the human pandemic H1N1 and H5N1. Our work demonstrates that the NA broad-inhibition precursor MBCs exist in healthy adults and could be targeted by the NA-based universal flu vaccine.
Collapse
Affiliation(s)
- Xin Wang
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Huihui Kong
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Bingxin Chu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Qian Yang
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Chaohui Lin
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Rui Liu
- School of Life Science, Westlake University, Hangzhou, Zhejiang, China
| | - Changxu Chen
- School of Life Science, Westlake University, Hangzhou, Zhejiang, China
| | - Yang Gao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Guojun Wang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Dayan Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; WHO Collaborating Center for Reference and Research on Influenza, Beijing, China
| | - Chen Qin
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xiaohua Ye
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Lifei Yu
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Xiangfei Xu
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Jie Jin
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Ren Sun
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; School of Life Science, Westlake University, Hangzhou, Zhejiang, China
| | - Hualan Chen
- State Key Laboratory for Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Xudong Wu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| | - Zeli Zhang
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; School of Life Science, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Bayarri-Olmos R, Sutta A, Rosbjerg A, Mortensen MM, Helgstrand C, Nielsen PF, Pérez-Alós L, González-García B, Johnsen LB, Matthiesen F, Egebjerg T, Hansen CB, Sette A, Grifoni A, da Silva Antunes R, Garred P. Unraveling the impact of SARS-CoV-2 mutations on immunity: insights from innate immune recognition to antibody and T cell responses. Front Immunol 2024; 15:1412873. [PMID: 39720734 PMCID: PMC11666439 DOI: 10.3389/fimmu.2024.1412873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 11/22/2024] [Indexed: 12/26/2024] Open
Abstract
Throughout the COVID-19 pandemic, the emergence of new viral variants has challenged public health efforts, often evading antibody responses generated by infections and vaccinations. This immune escape has led to waves of breakthrough infections, raising questions about the efficacy and durability of immune protection. Here we focus on the impact of SARS-CoV-2 Delta and Omicron spike mutations on ACE-2 receptor binding, protein stability, and immune response evasion. Delta and Omicron variants had 3-5 times higher binding affinities to ACE-2 than the ancestral strain (KDwt = 23.4 nM, KDDelta = 8.08 nM, KDBA.1 = 4.77 nM, KDBA.2 = 4.47 nM). The pattern recognition molecule mannose-binding lectin (MBL) has been shown to recognize the spike protein. Here we found that MBL binding remained largely unchanged across the variants, even after introducing mutations at single glycan sites. Although MBL binding decreased post-vaccination, it increased by 2.6-fold upon IgG depletion, suggesting a compensatory or redundant role in immune recognition. Notably, we identified two glycan sites (N717 and N801) as potentially essential for the structural integrity of the spike protein. We also evaluated the antibody and T cell responses. Neutralization by serum immunoglobulins was predominantly mediated by IgG rather than IgA and was markedly impaired against the Delta (5.8-fold decrease) and Omicron variants BA.1 (17.4-fold) and BA.2 (14.2-fold). T cell responses, initially conserved, waned rapidly within 3 months post-Omicron infection. Our data suggests that immune imprinting may have hindered antibody and T cell responses toward the variants. Overall, despite decreased antibody neutralization, MBL recognition and T cell responses were generally unaffected by the variants. These findings extend our understanding of the complex interplay between viral adaptation and immune response, underscoring the importance of considering MBL interactions, immune imprinting, and viral evolution dynamics in developing new vaccine and treatment strategies.
Collapse
Affiliation(s)
- Rafael Bayarri-Olmos
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Recombinant Protein and Antibody Unit, Copenhagen University Hospital,
Rigshospitalet, Copenhagen, Denmark
| | - Adrian Sutta
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Recombinant Protein and Antibody Unit, Copenhagen University Hospital,
Rigshospitalet, Copenhagen, Denmark
| | - Anne Rosbjerg
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Recombinant Protein and Antibody Unit, Copenhagen University Hospital,
Rigshospitalet, Copenhagen, Denmark
| | | | | | | | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Beatriz González-García
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | - Cecilie Bo Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, United States
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | | | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Espi M, Charmetant X, Benotmane I, Lefsihane K, Barateau V, Gallais F, Boulenouar H, Ovize A, Barbry A, Bouz C, Morelon E, Defrance T, Fafi-Kremer S, Caillard S, Thaunat O. Memory B Cells Provide Long-Term Protection to Vaccinated Kidney Transplant Recipients Against SARS-CoV-2 Variants. J Med Virol 2024; 96:e70037. [PMID: 39530340 DOI: 10.1002/jmv.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/10/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Kidney transplant recipients (KTRs) are highly vulnerable to COVID-19. An intensified scheme of vaccination offers short-term protection to the 50%-75% of KTRs able to develop a germinal center reaction, required for the generation of neutralizing titers of antibodies (NAbs). However, the duration of this vaccinal protection is unknown. In-depth longitudinal analysis of the immune response to vaccination of 33 KTRs demonstrates that the low peak of IgGs, the progressive decline in antibody titers, and the emergence of a variant of concerns (VOC) of SARS-CoV2, synergize to let 2/3 of responders to vaccine without NAbs after only a few months. Yet, a retrospective study of an independent cohort of 274 KTRs, revealed that the risk of severe COVID-19 in the latter was low, similar to that of patients with serum neutralizing capacity against VOC. Our work links this late vaccine protection with the presence of memory B cells, which are generated during the initial vaccine-induced germinal center reaction, have a wide repertoire directed against conserved spike epitopes, and rapidly differentiate into IgG-producing plasma cells upon antigenic rechallenge. We conclude that in contrast with a serological layer that goes fading rapidly, the cellular layer of humoral memory provides an efficient long-term protection against VOC to KTRs. This illustration of the complementary roles of the two layers of the humoral memory has implications in immunopathology beyond the COVID-19 in KTRs.
Collapse
Affiliation(s)
- Maxime Espi
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Xavier Charmetant
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
- Department of Transplantation, Hospices Civils de Lyon, Edouard Herriot Hospital, Nephrology and Clinical Immunology, Lyon, France
- Claude Bernard University, Villeurbanne, France
| | - Ilies Benotmane
- Department of Nephrology Dialysis and Transplantation, Strasbourg University Hospital, Strasbourg, France
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Katia Lefsihane
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Véronique Barateau
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Floriane Gallais
- Department of Virology, Strasbourg University Hospital, Strasbourg, France
| | - Hafsa Boulenouar
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Anne Ovize
- Eurofins Biomnis Laboratory, Lyon, France
| | | | | | - Emmanuel Morelon
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
- Department of Transplantation, Hospices Civils de Lyon, Edouard Herriot Hospital, Nephrology and Clinical Immunology, Lyon, France
- Claude Bernard University, Villeurbanne, France
| | - Thierry Defrance
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Samira Fafi-Kremer
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
- Department of Virology, Strasbourg University Hospital, Strasbourg, France
| | - Sophie Caillard
- Department of Nephrology Dialysis and Transplantation, Strasbourg University Hospital, Strasbourg, France
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Olivier Thaunat
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
- Department of Transplantation, Hospices Civils de Lyon, Edouard Herriot Hospital, Nephrology and Clinical Immunology, Lyon, France
- Claude Bernard University, Villeurbanne, France
| |
Collapse
|
6
|
Donadeu L, Gomez-Olles S, Casanova F, Torija A, Lopez-Meseguer M, Boada-Pérez M, Kervella D, Crespo E, Carrera-Muñoz C, Campos-Varela I, Castells L, Cortese MF, Esperalba J, Fernández-Naval C, Quintero J, Muñoz M, Agüero F, Gonzalez-Costello J, Lladó L, Favà A, Cañas L, del Mar de la Hoz-Caballero M, Meneghini M, Torres IB, Juvé M, Hafkamp FMJ, Vila M, Robles AG, Buzón MJ, Toapanta N, Zúñiga JM, Monforte V, Saez-Giménez B, Len O, Arcos IL, Miret E, Ariceta G, Pardo E, Martínez X, Moreso F, Bestard O. Role of SARS-CoV-2-specific memory B cells promoting immune protection after booster vaccination in solid organ transplantation. Front Immunol 2024; 15:1463769. [PMID: 39439787 PMCID: PMC11493670 DOI: 10.3389/fimmu.2024.1463769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Solid organ transplant (SOT) recipients display weak seroconversion and neutralizing antibody (NAb) responses after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination and remain at risk of severe coronavirus disease 2019 (COVID-19). While B-cell memory is the hallmark of serological immunity, its role in driving successful vaccine responses and providing immune protection in SOT patients remains unclear. Methods We investigated the function and interplay of SARS-CoV-2-specific memory B cells (mBc), different cytokineproducing T cells, and cross-reactive NAb in driving seroconversion and protection against COVID-19 in two cohorts. First, we studied a large cohort of 148 SOT recipients and 32 immunocompetent individuals who underwent several vaccinations. Subsequently, we assessed 25 SOT patients participating in a randomized controlled trial to compare two different immunosuppressive strategies for allowing successful seroconversion and memory-cell responses after booster vaccination. Results We corroborate previous findings that B- and T-cell memory responses are weaker and more delayed in SOT patients than in immunocompetent (IC) individuals; however, within the SOT cohort, we found that these responses are relatively stronger and more robust in patients not receiving mycophenolate mofetil (MMF)-based therapies. Anti- spike IgG titers strongly correlated with RBD-specific IgG-producing mBc, with both displaying broad viral cross reactivity. Prebooster SARS-CoV-2-specific mBc and IL-2- producing T cells accurately predicted Nab seroconversion (AUC, 0.828) and protection against severe COVID-19. While switching unresponsive SOT patients from calcineurin inhibitors (CNI)/MMF to a low-exposure CNI/mTOR-i regimen favored wider SARS-CoV-2-specific immune responses after a fourth booster vaccination, preformed RBD-specific mBc predicted NAb seroconversion. Discussion Our study adds new insights into the pathobiology of immune memory and highlights the pivotal role of SARS-CoV-2-specific mBc in promoting immune protection inSOT patients.
Collapse
Affiliation(s)
- Laura Donadeu
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Susana Gomez-Olles
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Franc Casanova
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba Torija
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Lopez-Meseguer
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Lung Transplant Unit, Pneumology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Meritxell Boada-Pérez
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Delphine Kervella
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elena Crespo
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Claudia Carrera-Muñoz
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Isabel Campos-Varela
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Liver Unit, Vall d'Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Lluís Castells
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Liver Unit, Vall d'Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria F. Cortese
- Microbiology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juliana Esperalba
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Microbiology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Candela Fernández-Naval
- Microbiology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jesús Quintero
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Pediatric Hepatology and Liver Transplant Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marina Muñoz
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Pediatric Nephrology, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Fernando Agüero
- Department of Preventive Medicine and Epidemiology, Bellvitge University Hospital, Barcelona, Spain
| | - José Gonzalez-Costello
- Advanced Heart Failure and Heart Transplant Unit, Department of Cardiology, Hospital Universitari de Bellvitge, BIOHEART-Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), Universitat de Barcelona, Ciber Cardiovascular (CIBERCV), Barcelona, Spain
| | - Laura Lladó
- Liver Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - Alexandre Favà
- Kidney Transplant Unit, Bellvitge University Hospital, Barcelona, Spain
| | - Laura Cañas
- Kidney Transplant Unit, Nephrology department, Germans Trias i Pujol Hospital, Badalona, Spain
| | - María del Mar de la Hoz-Caballero
- Equipo de Atención Primaria Sant Rafael, Servei d'Atenció Primària (SAP) Muntanya, Gerència Territorial de Barcelona Ciutat, Institut Català de la Salut, Barcelona, Spain
| | - Maria Meneghini
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Irina B. Torres
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mariona Juvé
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - FMJ Hafkamp
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta Vila
- Microbiology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba G. Robles
- Infectious Diseases Department, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria José Buzón
- Infectious Diseases Department, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Nestor Toapanta
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - José Miguel Zúñiga
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Víctor Monforte
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Lung Transplant Unit, Pneumology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Berta Saez-Giménez
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Laboratory of Pneumology, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Lung Transplant Unit, Pneumology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Oscar Len
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Infectious Diseases, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ibai Los Arcos
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Infectious Diseases, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Enric Miret
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Urology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gema Ariceta
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Pediatric Nephrology, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Emma Pardo
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Martínez
- Department of Preventive Medicine and Epidemiology, Vall d’Hebron Hospital Universitari, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francesc Moreso
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Oriol Bestard
- Laboratory of Nephrology and Transplantation, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Vall d’Hebron for Solid Organ Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Kidney Transplant Unit, Nephrology Department, Vall d’Hebron Hospital Universitari, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
7
|
Jeger-Madiot R, Planas D, Staropoli I, Debarnot H, Kervevan J, Mary H, Collina C, Fonseca BF, Robinot R, Gellenoncourt S, Schwartz O, Ewart L, Bscheider M, Gobaa S, Chakrabarti LA. Modeling memory B cell responses in a lymphoid organ-chip to evaluate mRNA vaccine boosting. J Exp Med 2024; 221:e20240289. [PMID: 39240335 PMCID: PMC11383861 DOI: 10.1084/jem.20240289] [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: 02/14/2024] [Revised: 06/26/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024] Open
Abstract
Predicting the immunogenicity of candidate vaccines in humans remains a challenge. To address this issue, we developed a lymphoid organ-chip (LO chip) model based on a microfluidic chip seeded with human PBMC at high density within a 3D collagen matrix. Perfusion of the SARS-CoV-2 spike protein mimicked a vaccine boost by inducing a massive amplification of spike-specific memory B cells, plasmablast differentiation, and spike-specific antibody secretion. Features of lymphoid tissue, including the formation of activated CD4+ T cell/B cell clusters and the emigration of matured plasmablasts, were recapitulated in the LO chip. Importantly, myeloid cells were competent at capturing and expressing mRNA vectored by lipid nanoparticles, enabling the assessment of responses to mRNA vaccines. Comparison of on-chip responses to Wuhan monovalent and Wuhan/Omicron bivalent mRNA vaccine boosts showed equivalent induction of Omicron neutralizing antibodies, pointing at immune imprinting as reported in vivo. The LO chip thus represents a versatile platform suited to the preclinical evaluation of vaccine-boosting strategies.
Collapse
Affiliation(s)
- Raphaël Jeger-Madiot
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Delphine Planas
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Isabelle Staropoli
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Hippolyte Debarnot
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Jérôme Kervevan
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Héloïse Mary
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Camilla Collina
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Barbara F. Fonseca
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Rémy Robinot
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Stacy Gellenoncourt
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Olivier Schwartz
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | | | - Michael Bscheider
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Samy Gobaa
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Lisa A. Chakrabarti
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| |
Collapse
|
8
|
Luo M, Zhou R, Tang B, Liu H, Chen B, Liu N, Mo Y, Zhang P, Lee YL, Ip JD, Wing-Ho Chu A, Chan WM, Man HO, Chen Y, To KKW, Yuen KY, Dang S, Chen Z. Ultrapotent class I neutralizing antibodies post Omicron breakthrough infection overcome broad SARS-CoV-2 escape variants. EBioMedicine 2024; 108:105354. [PMID: 39341153 PMCID: PMC11470419 DOI: 10.1016/j.ebiom.2024.105354] [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: 06/28/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The spread of emerging SARS-CoV-2 immune escape sublineages, especially JN.1 and KP.2, has resulted in new waves of COVID-19 globally. The evolving memory B cell responses elicited by the parental Omicron variants to subvariants with substantial antigenic drift remain incompletely investigated. METHODS Using the single B cell antibody cloning technology, we isolated single memory B cells, delineated the B cell receptor repertoire and conducted the pseudovirus-based assay for recovered neutralizing antibodies (NAb) screening. We analyzed the cryo-EM structures of top broadly NAbs (bnAbs) and evaluated their in vivo efficacy (golden Syrian hamster model). FINDINGS By investigating the evolution of human B cell immunity, we discovered a new panel of bnAbs arising from vaccinees after Omicron BA.2/BA.5 breakthrough infections. Two lead bnAbs neutralized major Omicron subvariants including JN.1 and KP.2 with IC50 values less than 10 ng/mL, representing ultrapotent receptor binding domain (RBD)-specific class I bnAbs. They belonged to the IGHV3-53/3-66 clonotypes instead of evolving from the pre-existing vaccine-induced IGHV1-58/IGKV3-20 bnAb ZCB11. Despite sequence diversity, they targeted previously unrecognized, highly conserved conformational epitopes in the receptor binding motif (RBM) for ultrapotent ACE2 blockade. The lead bnAb ZCP3B4 not only protected the lungs of hamsters intranasally challenged with BA.5.2, BQ.1.1 and XBB.1.5 but also prevented their contact transmission. INTERPRETATION Our findings demonstrated that class I bnAbs have evolved an ultrapotent mode of action protecting against highly transmissible and broad Omicron escape variants, and their epitopes are potential targets for novel bnAbs and vaccine development. FUNDING A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.
Collapse
Affiliation(s)
- Mengxiao Luo
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Runhong Zhou
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China
| | - Bingjie Tang
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, People's Republic of China
| | - Hang Liu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, People's Republic of China
| | - Bohao Chen
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Na Liu
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Yufei Mo
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Pengfei Zhang
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Ye Lim Lee
- Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Jonathan Daniel Ip
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Allen Wing-Ho Chu
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Wan-Mui Chan
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Hiu-On Man
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Yuting Chen
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Kelvin Kai-Wang To
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong SAR, People's Republic of China; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Kwok-Yung Yuen
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong SAR, People's Republic of China; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Shangyu Dang
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, People's Republic of China; HKUST-Shenzhen Research Institute, Nanshan, Shenzhen, 518057, People's Republic of China.
| | - Zhiwei Chen
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China; Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Health@InnoHK, The University of Hong Kong, Hong Kong SAR, People's Republic of China; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China.
| |
Collapse
|
9
|
Fryer HA, Geers D, Gommers L, Zaeck LM, Tan NH, Jones-Freeman B, Goorhuis A, Postma DF, Visser LG, Hogarth PM, Koopmans MPG, GeurtsvanKessel CH, O'Hehir RE, van der Kuy PHM, de Vries RD, van Zelm MC. Fourth dose bivalent COVID-19 vaccines outperform monovalent boosters in eliciting cross-reactive memory B cells to Omicron subvariants. J Infect 2024; 89:106246. [PMID: 39127451 DOI: 10.1016/j.jinf.2024.106246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Bivalent COVID-19 vaccines comprising ancestral Wuhan-Hu-1 (WH1) and the Omicron BA.1 or BA.5 subvariant elicit enhanced serum antibody responses to emerging Omicron subvariants. Here, we characterized the RBD-specific memory B cell (Bmem) response following a fourth dose with a BA.1 or BA.5 bivalent vaccine, in direct comparison with a WH1 monovalent fourth dose. Healthcare workers previously immunized with mRNA or adenoviral vector monovalent vaccines were sampled before and one month after a fourth dose with a monovalent or a BA.1 or BA.5 bivalent vaccine. Serum neutralizing antibodies (NAb) were quantified, as well as RBD-specific Bmem with an in-depth spectral flow cytometry panel including recombinant RBD proteins of the WH1, BA.1, BA.5, BQ.1.1, and XBB.1.5 variants. Both bivalent vaccines elicited higher NAb titers against Omicron subvariants compared to the monovalent vaccine. Following either vaccine type, recipients had slightly increased WH1 RBD-specific Bmem numbers. Both bivalent vaccines significantly increased WH1 RBD-specific Bmem binding of all Omicron subvariants tested by flow cytometry, while recognition of Omicron subvariants was not enhanced following monovalent vaccination. IgG1+ Bmem dominated the response, with substantial IgG4+ Bmem only detected in recipients of an mRNA vaccine for their primary dose. Thus, Omicron-based bivalent vaccines can significantly boost NAb and Bmem specific for ancestral WH1 and Omicron variants and improve recognition of descendent subvariants by pre-existing, WH1-specific Bmem beyond that of a monovalent vaccine. This provides new insights into the capacity of variant-based mRNA booster vaccines to improve immune memory against emerging SARS-CoV-2 variants and potentially protect against severe disease. ONE-SENTENCE SUMMARY: Omicron BA.1 and BA.5 bivalent COVID-19 boosters, used as a fourth dose, increase RBD-specific Bmem cross-recognition of Omicron subvariants, both those encoded by the vaccines and antigenically distinct subvariants, further than a monovalent booster.
Collapse
Affiliation(s)
- Holly A Fryer
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Daryl Geers
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Lennert Gommers
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Luca M Zaeck
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Ngoc H Tan
- Dept. Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Bernadette Jones-Freeman
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Abraham Goorhuis
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Centers, Amsterdam, the Netherlands; Infection and Immunity, Amsterdam Public Health, University of Amsterdam, Amsterdam, the Netherlands
| | - Douwe F Postma
- Department of Internal Medicine and Infectious Diseases, University Medical Center Groningen, Groningen, the Netherlands
| | - Leo G Visser
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - P Mark Hogarth
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Immune Therapies Group, Burnet Institute, Melbourne, Victoria, Australia
| | - Marion P G Koopmans
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | | | - Robyn E O'Hehir
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, Victoria, Australia
| | - P Hugo M van der Kuy
- Dept. Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Rory D de Vries
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Menno C van Zelm
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, Victoria, Australia; Dept. Immunology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
10
|
Dejoux A, Zhu Q, Ganneau C, Goff ORL, Godon O, Lemaitre J, Relouzat F, Huetz F, Sokal A, Vandenberghe A, Pecalvel C, Hunault L, Derenne T, Gillis CM, Iannascoli B, Wang Y, Rose T, Mertens C, Nicaise-Roland P, England P, Mahévas M, de Chaisemartin L, Le Grand R, Letscher H, Saul F, Pissis C, Haouz A, Reber LL, Chappert P, Jönsson F, Ebo DG, Millot GA, Bay S, Chollet-Martin S, Gouel-Chéron A, Bruhns P. Rocuronium-specific antibodies drive perioperative anaphylaxis but can also function as reversal agents in preclinical models. Sci Transl Med 2024; 16:eado4463. [PMID: 39259810 DOI: 10.1126/scitranslmed.ado4463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/06/2024] [Accepted: 08/05/2024] [Indexed: 09/13/2024]
Abstract
Neuromuscular blocking agents (NMBAs) relax skeletal muscles to facilitate surgeries and ease intubation but can lead to adverse reactions, including complications because of postoperative residual neuromuscular blockade (rNMB) and, in rare cases, anaphylaxis. Both adverse reactions vary between types of NMBAs, with rocuronium, a widely used nondepolarizing NMBA, inducing one of the longest rNMB durations and highest anaphylaxis incidences. rNMB induced by rocuronium can be reversed by the synthetic γ-cyclodextrin sugammadex. However, in rare cases, sugammadex can provoke anaphylaxis. Thus, additional therapeutic options are needed. Rocuronium-induced anaphylaxis is proposed to rely on preexisting rocuronium-binding antibodies. To understand the pathogenesis of rocuronium-induced anaphylaxis and to identify potential therapeutics, we investigated the memory B cell antibody repertoire of patients with suspected hypersensitivity to rocuronium. We identified polyclonal antibody repertoires with a high diversity among V(D)J genes without evidence of clonal groups. When recombinantly expressed, these antibodies demonstrated specificity and low affinity for rocuronium without cross-reactivity for other NMBAs. Moreover, when these antibodies were expressed as human immunoglobulin E (IgE), they triggered human mast cell activation and passive systemic anaphylaxis in transgenic mice, although their affinities were insufficient to serve as reversal agents. Rocuronium-specific, high-affinity antibodies were thus isolated from rocuronium-immunized mice. The highest-affinity antibody was able to reverse rocuronium-induced neuromuscular blockade in nonhuman primates with kinetics comparable to that of sugammadex. Together, these data support the hypothesis that antibodies cause anaphylactic reactions to rocuronium and pave the way for improved diagnostics and neuromuscular blockade reversal agents.
Collapse
Affiliation(s)
- Alice Dejoux
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Sorbonne Université, Collège Doctoral, 75005 Paris, France
| | - Qianqian Zhu
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Université Paris-Saclay, INSERM, Inflammation Microbiome Immunosurveillance, 91400 Orsay, France
| | - Christelle Ganneau
- Institut Pasteur, Université Paris Cité, CNRS UMR3523, Chimie des Biomolécules, 75015 Paris, France
| | - Odile Richard-Le Goff
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Ophélie Godon
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Julien Lemaitre
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases, 92260 Fontenay-aux-Roses and 94250 Le Kremlin-Bicêtre, France
| | - Francis Relouzat
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases, 92260 Fontenay-aux-Roses and 94250 Le Kremlin-Bicêtre, France
| | - François Huetz
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Aurélien Sokal
- Institut Necker Enfants Malades, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, Université Paris Cité, Université Paris Est-Créteil, 94000 Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale, Université Paris-Est Créteil, 94000 Créteil, France
- Service de Médecine interne, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris Cité, 92110 Clichy, France
| | - Alexis Vandenberghe
- Institut Necker Enfants Malades, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, Université Paris Cité, Université Paris Est-Créteil, 94000 Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale, Université Paris-Est Créteil, 94000 Créteil, France
| | - Cyprien Pecalvel
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, 31000 Toulouse, France
| | - Lise Hunault
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Sorbonne Université, Collège Doctoral, 75005 Paris, France
| | - Thomas Derenne
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Sorbonne Université, Collège Doctoral, 75005 Paris, France
| | - Caitlin M Gillis
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Bruno Iannascoli
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Yidan Wang
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Thierry Rose
- Institut Pasteur, Université Paris Cité, INSERM UMR1224, Biologie Cellulaire des Lymphocytes, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, 75015 Paris, France
| | - Christel Mertens
- Faculty of Medicine and Health Science, Department of Immunology-Allergology-Rheumatology, Antwerp University Hospital and the Infla-Med Center of Excellence, University of Antwerp, Antwerp, Belgium; Department of Immunology and Allergology, AZ Jan Palfijn Ghent, 9000 Ghent, Belgium
| | - Pascale Nicaise-Roland
- Service d'immunologie Biologique, DMU BIOGEM, Hôpital Bichat, APHP, 75018, Paris, France
| | - Patrick England
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Molecular Biophysics Core Facility, 75015 Paris, France
| | - Matthieu Mahévas
- Institut Necker Enfants Malades, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, Université Paris Cité, Université Paris Est-Créteil, 94000 Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale, Université Paris-Est Créteil, 94000 Créteil, France
| | - Luc de Chaisemartin
- Université Paris-Saclay, INSERM, Inflammation Microbiome Immunosurveillance, 91400 Orsay, France
- Service d'immunologie Biologique, DMU BIOGEM, Hôpital Bichat, APHP, 75018, Paris, France
| | - Roger Le Grand
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases, 92260 Fontenay-aux-Roses and 94250 Le Kremlin-Bicêtre, France
| | - Hélène Letscher
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases, 92260 Fontenay-aux-Roses and 94250 Le Kremlin-Bicêtre, France
| | - Frederick Saul
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Plate-forme Cristallographie-C2RT, 75015 Paris, France
| | - Cédric Pissis
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Plate-forme Cristallographie-C2RT, 75015 Paris, France
| | - Ahmed Haouz
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Plate-forme Cristallographie-C2RT, 75015 Paris, France
| | - Laurent L Reber
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, 31000 Toulouse, France
| | - Pascal Chappert
- Institut Necker Enfants Malades, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, Université Paris Cité, Université Paris Est-Créteil, 94000 Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale, Université Paris-Est Créteil, 94000 Créteil, France
| | - Friederike Jönsson
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- CNRS, F-75015 Paris, France
| | - Didier G Ebo
- Faculty of Medicine and Health Science, Department of Immunology-Allergology-Rheumatology, Antwerp University Hospital and the Infla-Med Center of Excellence, University of Antwerp, Antwerp, Belgium; Department of Immunology and Allergology, AZ Jan Palfijn Ghent, 9000 Ghent, Belgium
| | - Gaël A Millot
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, 75015 Paris, France
| | - Sylvie Bay
- Institut Pasteur, Université Paris Cité, CNRS UMR3523, Chimie des Biomolécules, 75015 Paris, France
| | - Sylvie Chollet-Martin
- Université Paris-Saclay, INSERM, Inflammation Microbiome Immunosurveillance, 91400 Orsay, France
- Service d'immunologie Biologique, DMU BIOGEM, Hôpital Bichat, APHP, 75018, Paris, France
| | - Aurélie Gouel-Chéron
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Anaesthesiology and Critical Care Medicine Department, DMU Parabol, Bichat-Claude Bernard Hospital, AP-HP, 75018 Paris, France
- Université Paris Cité, 75010 Paris, France
| | - Pierre Bruhns
- Institut Pasteur, Université Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- INSERM 1152, DHU FIRE, Labex Inflamex, Université Paris Diderot Paris 7, 75018 Paris, France
| |
Collapse
|
11
|
Wang M, Zhang D, Lei T, Zhou Y, Qin H, Wu Y, Liu S, Zhang L, Jia K, Dong Y, Wang S, Li Y, Fan Y, Gui L, Dong Y, Zhang W, Li Z, Hou J. Interferon-responsive neutrophils and macrophages extricate SARS-CoV-2 Omicron critical patients from the nasty fate of sepsis. J Med Virol 2024; 96:e29889. [PMID: 39206862 DOI: 10.1002/jmv.29889] [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: 04/22/2024] [Revised: 07/24/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
The SARS-CoV-2 Omicron variant is characterized by its high transmissibility, which has caused a worldwide epidemiological event. Yet, it turns ominous once the disease progression degenerates into severe pneumonia and sepsis, presenting a horrendous lethality. To elucidate the alveolar immune or inflammatory landscapes of Omicron critical-ill patients, we performed single-cell RNA-sequencing (scRNA-seq) of bronchoalveolar lavage fluid (BALF) from the patients with critical pneumonia caused by Omicron infection, and analyzed the correlation between the clinical severity scores and different immune cell subpopulations. In the BALF of Omicron critical patients, the alveolar violent myeloid inflammatory environment was determined. ISG15+ neutrophils and CXCL10+ macrophages, both expressed the interferon-stimulated genes (ISGs), were negatively correlated with clinical pulmonary infection score, while septic CST7+ neutrophils and inflammatory VCAN+ macrophages were positively correlated with sequential organ failure assessment. The percentages of ISG15+ neutrophils were associated with more protective alveolar epithelial cells, and may reshape CD4+ T cells to the exhaustive phenotype, thus preventing immune injuries. The CXCL10+ macrophages may promote plasmablast/plasma cell survival and activation as well as the production of specific antibodies. As compared to the previous BALF scRNA-seq data from SARS-CoV-2 wild-type/Alpha critical patients, the subsets of neutrophils and macrophages with pro-inflammatory and immunoregulatory features presented obvious distinctions, suggesting an immune disparity in Omicron variants. Overall, this study provides a BALF single-cell atlas of Omicron critical patients, and suggests that alveolar interferon-responsive neutrophils and macrophages may extricate SARS-CoV-2 Omicron critical patients from the nasty fate of sepsis.
Collapse
Affiliation(s)
- Mu Wang
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Dingji Zhang
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Ting Lei
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Ye Zhou
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Hao Qin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Yanfeng Wu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Shuxun Liu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Liyuan Zhang
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Kaiwei Jia
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Yue Dong
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Suyuan Wang
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Yunhui Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Yiwen Fan
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Liangchen Gui
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Yuchao Dong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Zhixuan Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Jin Hou
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| |
Collapse
|
12
|
Tarke A, Ramezani-Rad P, Alves Pereira Neto T, Lee Y, Silva-Moraes V, Goodwin B, Bloom N, Siddiqui L, Avalos L, Frazier A, Zhang Z, da Silva Antunes R, Dan J, Crotty S, Grifoni A, Sette A. SARS-CoV-2 breakthrough infections enhance T cell response magnitude, breadth, and epitope repertoire. Cell Rep Med 2024; 5:101583. [PMID: 38781962 PMCID: PMC11228552 DOI: 10.1016/j.xcrm.2024.101583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/22/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
Little is known about the effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 or SARS2) vaccine breakthrough infections (BTIs) on the magnitude and breadth of the T cell repertoire after exposure to different variants. We studied samples from individuals who experienced symptomatic BTIs during Delta or Omicron waves. In the pre-BTI samples, 30% of the donors exhibited substantial immune memory against non-S (spike) SARS2 antigens, consistent with previous undiagnosed asymptomatic SARS2 infections. Following symptomatic BTI, we observed (1) enhanced S-specific CD4 and CD8 T cell responses in donors without previous asymptomatic infection, (2) expansion of CD4 and CD8 T cell responses to non-S targets (M, N, and nsps) independent of SARS2 variant, and (3) generation of novel epitopes recognizing variant-specific mutations. These variant-specific T cell responses accounted for 9%-15% of the total epitope repertoire. Overall, BTIs boost vaccine-induced immune responses by increasing the magnitude and by broadening the repertoire of T cell antigens and epitopes recognized.
Collapse
Affiliation(s)
- Alison Tarke
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Parham Ramezani-Rad
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | | | - Yeji Lee
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Vanessa Silva-Moraes
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
| | - Benjamin Goodwin
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Nathaniel Bloom
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Leila Siddiqui
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Liliana Avalos
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - April Frazier
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Zeli Zhang
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | | | - Jennifer Dan
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Shane Crotty
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA.
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| |
Collapse
|
13
|
Wang X, Zhang M, Wei K, Li C, Yang J, Jiang S, Zhao C, Zhao X, Qiao R, Cui Y, Chen Y, Li J, Cai G, Liu C, Yu J, Zhang W, Xie F, Wang P, Zhang Y. Longitudinal Analysis of Humoral and Cellular Immune Response up to 6 Months after SARS-CoV-2 BA.5/BF.7/XBB Breakthrough Infection and BA.5/BF.7-XBB Reinfection. Vaccines (Basel) 2024; 12:464. [PMID: 38793715 PMCID: PMC11125724 DOI: 10.3390/vaccines12050464] [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: 03/25/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
The rapid mutation of SARS-CoV-2 has led to multiple rounds of large-scale breakthrough infection and reinfection worldwide. However, the dynamic changes of humoral and cellular immunity responses to several subvariants after infection remain unclear. In our study, a 6-month longitudinal immune response evaluation was conducted on 118 sera and 50 PBMC samples from 49 healthy individuals who experienced BA.5/BF.7/XBB breakthrough infection or BA.5/BF.7-XBB reinfection. By studying antibody response, memory B cell, and IFN-γ secreting CD4+/CD8+ T cell response to several SARS-CoV-2 variants, we observed that each component of immune response exhibited distinct kinetics. Either BA.5/BF.7/XBB breakthrough infection or BA.5/BF.7-XBB reinfection induces relatively high level of binding and neutralizing antibody titers against Omicron subvariants at an early time point, which rapidly decreases over time. Most of the individuals at 6 months post-breakthrough infection completely lost their neutralizing activities against BQ.1.1, CH.1.1, BA.2.86, JN.1 and XBB subvariants. Individuals with BA.5/BF.7-XBB reinfection exhibit immune imprinting shifting and recall pre-existing BA.5/BF.7 neutralization antibodies. In the BA.5 breakthrough infection group, the frequency of BA.5 and XBB.1.16-RBD specific memory B cells, resting memory B cells, and intermediate memory B cells gradually increased over time. On the other hand, the frequency of IFN-γ secreting CD4+/CD8+ T cells induced by WT/BA.5/XBB.1.16 spike trimer remains stable over time. Overall, our research indicates that individuals with breakthrough infection have rapidly declining antibody levels but have a relatively stable cellular immunity that can provide some degree of protection from future exposure to new antigens.
Collapse
Affiliation(s)
- Xun Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Meng Zhang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| | - Kaifeng Wei
- College of Traditional Chinese Medicine·College of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Chen Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Jinghui Yang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| | - Shujun Jiang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| | - Chaoyue Zhao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Xiaoyu Zhao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Rui Qiao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Yuchen Cui
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Yanjia Chen
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Jiayan Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Guonan Cai
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Changyi Liu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Jizhen Yu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Wenhong Zhang
- Department of Infectious Diseases, National Medical Center for Infectious Diseases and Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai 200437, China;
| | - Faren Xie
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| | - Pengfei Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200437, China; (X.W.); (C.L.); (C.Z.); (X.Z.); (R.Q.); (Y.C.); (Y.C.); (J.L.); (G.C.); (C.L.); (J.Y.)
| | - Yanliang Zhang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.Z.); (J.Y.); (S.J.)
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing 210001, China
| |
Collapse
|
14
|
Nantel S, Sheikh-Mohamed S, Chao GYC, Kurtesi A, Hu Q, Wood H, Colwill K, Li Z, Liu Y, Seifried L, Bourdin B, McGeer A, Hardy WR, Rojas OL, Al-Aubodah TA, Liu Z, Ostrowski MA, Brockman MA, Piccirillo CA, Quach C, Rini JM, Gingras AC, Decaluwe H, Gommerman JL. Comparison of Omicron breakthrough infection versus monovalent SARS-CoV-2 intramuscular booster reveals differences in mucosal and systemic humoral immunity. Mucosal Immunol 2024; 17:201-210. [PMID: 38278415 DOI: 10.1016/j.mucimm.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/17/2024] [Accepted: 01/20/2024] [Indexed: 01/28/2024]
Abstract
Our understanding of the quality of cellular and humoral immunity conferred by COVID-19 vaccination alone versus vaccination plus SARS-CoV-2 breakthrough (BT) infection remains incomplete. While the current (2023) SARS-CoV-2 immune landscape of Canadians is complex, in late 2021 most Canadians had either just received a third dose of COVID-19 vaccine, or had received their two-dose primary series and then experienced an Omicron BT. Herein we took advantage of this coincident timing to contrast cellular and humoral immunity conferred by three doses of vaccine versus two doses plus BT. Our results show thatBT infection induces cell-mediated immune responses to variants comparable to an intramuscular vaccine booster dose. In contrast, BT subjects had higher salivary immunoglobulin (Ig)G and IgA levels against the Omicron spike and enhanced reactivity to the ancestral spike for the IgA isotype, which also reacted with SARS-CoV-1. Serumneutralizing antibody levels against the ancestral strain and the variants were also higher after BT infection. Our results support the need for the development of intranasal vaccines that could emulate the enhanced mucosal and humoral immunity induced by Omicron BT without exposing individuals to the risks associated with SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Sabryna Nantel
- Sainte-Justine University Hospital and Research Center, Montréal, Québec, Canada; Microbiology, Infectiology and Immunology Department, Faculty of Medicine, University of Montréal, Montréal, Québec, Canada
| | | | - Gary Y C Chao
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Alexandra Kurtesi
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Queenie Hu
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Heidi Wood
- One Health Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Zhijie Li
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Ying Liu
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Laurie Seifried
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Benoîte Bourdin
- Sainte-Justine University Hospital and Research Center, Montréal, Québec, Canada
| | - Allison McGeer
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - William R Hardy
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Olga L Rojas
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Tho-Alfakar Al-Aubodah
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada; Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Zhiyang Liu
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada; Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Mario A Ostrowski
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Mark A Brockman
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada; Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Caroline Quach
- Sainte-Justine University Hospital and Research Center, Montréal, Québec, Canada; Microbiology, Infectiology and Immunology Department, Faculty of Medicine, University of Montréal, Montréal, Québec, Canada
| | - James M Rini
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Hélène Decaluwe
- Sainte-Justine University Hospital and Research Center, Montréal, Québec, Canada; Microbiology, Infectiology and Immunology Department, Faculty of Medicine, University of Montréal, Montréal, Québec, Canada; Pediatric Immunology and Rheumatology Division, Department of Pediatrics, University of Montréal, Montréal, Québec, Canada.
| | | |
Collapse
|
15
|
Wang X, Jiang S, Ma W, Li X, Wei K, Xie F, Zhao C, Zhao X, Wang S, Li C, Qiao R, Cui Y, Chen Y, Li J, Cai G, Liu C, Yu J, Li J, Hu Z, Zhang W, Jiang S, Li M, Zhang Y, Wang P. Enhanced neutralization of SARS-CoV-2 variant BA.2.86 and XBB sub-lineages by a tetravalent COVID-19 vaccine booster. Cell Host Microbe 2024; 32:25-34.e5. [PMID: 38029742 DOI: 10.1016/j.chom.2023.11.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/23/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023]
Abstract
Emerging SARS-CoV-2 sub-lineages like XBB.1.5, XBB.1.16, EG.5, HK.3 (FLip), and XBB.2.3 and the variant BA.2.86 have recently been identified. Understanding the efficacy of current vaccines on these emerging variants is critical. We evaluate the serum neutralization activities of participants who received COVID-19 inactivated vaccine (CoronaVac), those who received the recently approved tetravalent protein vaccine (SCTV01E), or those who had contracted a breakthrough infection with BA.5/BF.7/XBB virus. Neutralization profiles against a broad panel of 30 sub-lineages reveal that BQ.1.1, CH.1.1, and all the XBB sub-lineages exhibit heightened resistance to neutralization compared to previous variants. However, despite their extra mutations, BA.2.86 and the emerging XBB sub-lineages do not demonstrate significantly increased resistance to neutralization over XBB.1.5. Encouragingly, the SCTV01E booster consistently induces higher neutralizing titers against all these variants than breakthrough infection does. Cellular immunity assays also show that the SCTV01E booster elicits a higher frequency of virus-specific memory B cells. Our findings support the development of multivalent vaccines to combat future variants.
Collapse
Affiliation(s)
- Xun Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Shujun Jiang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing, Jiangsu, China
| | - Wentai Ma
- Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xiangnan Li
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, China
| | - Kaifeng Wei
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Faren Xie
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing, Jiangsu, China
| | - Chaoyue Zhao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaoyu Zhao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Shidi Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Chen Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Rui Qiao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yuchen Cui
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yanjia Chen
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Jiayan Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Guonan Cai
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Changyi Liu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Jizhen Yu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Jixi Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China
| | - Zixin Hu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, China; Artificial Intelligence Innovation and Incubation Institute, Fudan University, Shanghai, China
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Mingkun Li
- Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Yanliang Zhang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing, Jiangsu, China.
| | - Pengfei Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|