1
|
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
|
2
|
Nguyen HC, Lal KG, Balinsky CA, Hontz RD, Lin J, Beye MJ, Smith L, Pan L, Cheng Y, Fox I, Lizewski SE, Foo HS, Krebs SJ, Sun P, Letizia AG. Informing the Need for a SARS-CoV-2 Booster Based on the Immune Responses Among Young Healthy Adults to Variants Circulating in Late 2023. J Infect Dis 2024; 230:645-656. [PMID: 38718223 DOI: 10.1093/infdis/jiae249] [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: 02/14/2024] [Accepted: 05/07/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND COVID-19 remains a global public health challenge due to new immune-evasive SARS-CoV-2 variants and heterogeneous immunity. METHODS In this cross-sectional study, we evaluated the adaptive immune responses in US active duty personnel who completed a COVID-19 primary vaccine series and had heterogenous SARS-CoV-2 vaccination and infection histories to 3 previously dominant variants (ancestral, Delta, BA.5) and 3 circulating variants (XBB.1.5, EG.5, and BA.2.86) in late 2023. Analyses were based on the most recent exposure in terms of timing (within or beyond 12 months) and type (vaccine or infection). RESULTS Significant reduction was observed in binding antibodies, neutralization antibodies, memory B cells, and CD8+ T cells against circulating variants when compared with previous variants. The reduction in antibody response was more pronounced in those whose most recent exposure was >12 months from enrollment. In contrast, the CD4+ T-cell response was largely consistent across all tested variants. The type of most recent exposure was not a significant factor in determining the magnitude of current immune responses. CONCLUSIONS Administration of the XBB.1.5-based booster is likely to enhance cross-reactive humoral responses against SARS-CoV-2 circulating lineages. Ongoing surveillance of immune responses to emerging variants is needed for informing vaccine composition and timing.
Collapse
Affiliation(s)
- Huy C Nguyen
- US Naval Medical Research Unit INDO PACIFIC, Science Directorate, Singapore, Singapore
| | - Kerri G Lal
- Walter Reed Army Institute of Research, US Military HIV Research Program, B Cell Biology, Silver Spring
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | - Corey A Balinsky
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
- Naval Medical Research Command, Diagnostics Surveillance Division, Silver Spring
| | - Robert D Hontz
- US Naval Medical Research Unit INDO PACIFIC, Science Directorate, Singapore, Singapore
| | - Jin Lin
- US Naval Medical Research Unit INDO PACIFIC, Science Directorate, Singapore, Singapore
| | - Matthew J Beye
- US Naval Medical Research Unit INDO PACIFIC, Science Directorate, Singapore, Singapore
| | - Lauren Smith
- Walter Reed Army Institute of Research, US Military HIV Research Program, B Cell Biology, Silver Spring
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | - Li Pan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | | | - Isabella Fox
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | - Stephen E Lizewski
- Naval Medical Research Command, Diagnostics Surveillance Division, Silver Spring
| | - Hayley S Foo
- US Naval Medical Research Unit INDO PACIFIC, Science Directorate, Singapore, Singapore
| | - Shelly J Krebs
- Walter Reed Army Institute of Research, US Military HIV Research Program, B Cell Biology, Silver Spring
| | - Peifang Sun
- Naval Medical Research Command, Diagnostics Surveillance Division, Silver Spring
| | - Andrew G Letizia
- US Naval Medical Research Unit INDO PACIFIC, Science Directorate, Singapore, Singapore
| |
Collapse
|
3
|
Manfroi B, Cuc BT, Sokal A, Vandenberghe A, Temmam S, Attia M, El Behi M, Camaglia F, Nguyen NT, Pohar J, Salem-Wehbe L, Pottez-Jouatte V, Borzakian S, Elenga N, Galeotti C, Morelle G, de Truchis de Lays C, Semeraro M, Romain AS, Aubart M, Ouldali N, Mahuteau-Betzer F, Beauvineau C, Amouyal E, Berthaud R, Crétolle C, Arnould MD, Faye A, Lorrot M, Benoist G, Briand N, Courbebaisse M, Martin R, Van Endert P, Hulot JS, Blanchard A, Tartour E, Leite-de-Moraes M, Lezmi G, Ménager M, Luka M, Reynaud CA, Weill JC, Languille L, Michel M, Chappert P, Mora T, Walczak AM, Eloit M, Bacher P, Scheffold A, Mahévas M, Sermet-Gaudelus I, Fillatreau S. Preschool-age children maintain a distinct memory CD4 + T cell and memory B cell response after SARS-CoV-2 infection. Sci Transl Med 2024; 16:eadl1997. [PMID: 39292802 DOI: 10.1126/scitranslmed.adl1997] [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: 10/04/2023] [Accepted: 07/19/2024] [Indexed: 09/20/2024]
Abstract
The development of the human immune system lasts for several years after birth. The impact of this maturation phase on the quality of adaptive immunity and the acquisition of immunological memory after infection at a young age remains incompletely defined. Here, using an antigen-reactive T cell (ARTE) assay and multidimensional flow cytometry, we profiled circulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-reactive CD3+CD4+CD154+ T cells in children and adults before infection, during infection, and 11 months after infection, stratifying children into separate age groups and adults according to disease severity. During SARS-CoV-2 infection, children younger than 5 years old displayed a lower antiviral CD4+ T cell response, whereas children older than 5 years and adults with mild disease had, quantitatively and phenotypically, comparable virus-reactive CD4+ T cell responses. Adults with severe disease mounted a response characterized by higher frequencies of virus-reactive proinflammatory and cytotoxic T cells. After SARS-CoV-2 infection, preschool-age children not only maintained neutralizing SARS-CoV-2-reactive antibodies postinfection comparable to adults but also had phenotypically distinct memory T cells displaying high inflammatory features and properties associated with migration toward inflamed sites. Moreover, preschool-age children had markedly fewer circulating virus-reactive memory B cells compared with the other cohorts. Collectively, our results reveal unique facets of antiviral immunity in humans at a young age and indicate that the maturation of adaptive responses against SARS-CoV-2 toward an adult-like profile occurs in a progressive manner.
Collapse
Affiliation(s)
- Benoît Manfroi
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Bui Thi Cuc
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Aurélien Sokal
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
- Service de Médecine interne, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris (AP-HP), 92110 Clichy, France
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Alexis Vandenberghe
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
- INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Sarah Temmam
- Pathogen Discovery Laboratory, Institut Pasteur, Université Paris Cité, and Institut Pasteur, the WOAH Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Université Paris Cité, 75015 Paris, France
| | - Mikaël Attia
- Molecular Genetics of RNA Viruses, Department of Virology, Institut Pasteur, Université Paris-Cité, CNRS UMR 3569, 75015 Paris, France
| | - Mohamed El Behi
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Francesco Camaglia
- Laboratoire de physique de l'École normale supérieure, CNRS, Paris Sciences et Lettres (PSL) University, Sorbonne Université, and Université de Paris, 75005 Paris, France
| | - Ngan Thu Nguyen
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Jelka Pohar
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Immunology and Cellular Immunotherapy (ICI) Group, Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia
| | - Layale Salem-Wehbe
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Valentine Pottez-Jouatte
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Sibyline Borzakian
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- CNRS UMR 9187, INSERM U1196, Chemistry and Modeling for the Biological of Cancer, Institut Curie, PSL Research University, 91405 Orsay, France
- Université Paris-Saclay, 91405 Orsay, France
| | - Narcisse Elenga
- Service de Pédiatrie, Centre Hospitalier de Cayenne, 97300 French Guiana
| | - Caroline Galeotti
- Department of Pediatric Rheumatology, Bicêtre Hospital, AP-HP, Paris-Saclay University, 94275 Le Kremlin-Bicêtre, France
| | - Guillaume Morelle
- Department of General Paediatrics, Hôpital Bicêtre, AP-HP, University of Paris Saclay, 94275 Le Kremlin-Bicêtre, France
| | - Camille de Truchis de Lays
- Service de Pédiatrie. Hôpital Jean-Verdier, AP-HP, Hôpitaux Universitaires Paris Seine-Saint-Denis, 93140 Bondy, France
| | - Michaela Semeraro
- University of Paris Cité, and Clinical Investigation Center, Clinical Research Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Anne-Sophie Romain
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Trousseau Hospital, General Paediatrics Department, 75012 Paris, France
| | - Mélodie Aubart
- INSERM U1163, Genetic Predisposition to Infectious Diseases, Imagine Institute, Université Paris Cité, Paris F-75015, France
- Pediatric Neurology Department, Necker-Enfants Malades Universitary Hospital, AP-HP, Paris-Cité University, 75015 Paris, France
| | - Naim Ouldali
- Department of General Pediatrics, Pediatric Infectious Disease and Internal Medicine, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, 75019 Paris, France
- Paris Cité University, INSERM UMR 1137, Infection, Antimicrobials, Modelling, Evolution (IAME), 75018 Paris, France
| | - Florence Mahuteau-Betzer
- CNRS UMR 9187, INSERM U1196, Chemistry and Modeling for the Biological of Cancer, Institut Curie, PSL Research University, 91405 Orsay, France
- Université Paris-Saclay, 91405 Orsay, France
| | - Claire Beauvineau
- CNRS UMR 9187, INSERM U1196, Chemistry and Modeling for the Biological of Cancer, Institut Curie, PSL Research University, 91405 Orsay, France
- Université Paris-Saclay, 91405 Orsay, France
| | - Elsa Amouyal
- SIREDO Pediatric Oncology Center, Institut Curie, Paris-Science Lettres University, 75005 Paris, France
| | - Romain Berthaud
- Pediatric Nephrology, Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA) Reference Center, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
- Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France
| | - Célia Crétolle
- Département de Pédiatrie, Service de Chirurgie viscérale pédiatrique, Hôpital Universitaire Necker-Enfants Malades, GH Paris Centre, 75015 Paris, France
| | - Marc Duval Arnould
- Department of General Paediatrics, Hôpital Bicêtre, AP-HP, University of Paris Saclay, 94275 Le Kremlin-Bicêtre, France
| | - Albert Faye
- Pediatric Neurology Department, Necker-Enfants Malades Universitary Hospital, AP-HP, Paris-Cité University, 75015 Paris, France
| | - Mathie Lorrot
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Trousseau Hospital, General Paediatrics Department, 75012 Paris, France
| | - Grégoire Benoist
- Service de pédiatrie générale et hôpital de jour allergologie, CHU Ambroise-Paré, AP-HP, 92100 Boulogne-Billancourt, France
| | - Nelly Briand
- University of Paris Cité, and Clinical Investigation Center, Clinical Research Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Marie Courbebaisse
- Faculté de Médecine, Université Paris Cité, 75015 Paris, France
- Explorations fonctionnelles rénales, Physiologie, Hôpital européen Georges-Pompidou, Assistance Publique-Hôpitaux de Paris, 75908 Paris Cedex 15, France
| | - Roland Martin
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Therapeutic Immune Design, Center for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, 171 76 Stockholm, Sweden
| | - Peter Van Endert
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Service Immunologie Biologique, AP-HP, Hôpital Universitaire Necker-Enfants Malades, F-75015 Paris, France
| | - Jean-Sébastien Hulot
- PARCC, INSERM, Université Paris Cité, 75015 Paris, France
- Centre d'Investigation Clinique, AP-HP, INSERM CIC-1418, Européen Georges Pompidou Hospital, 75015 Paris, France
| | - Anne Blanchard
- Centre d'Investigation Clinique, AP-HP, INSERM CIC-1418, Européen Georges Pompidou Hospital, 75015 Paris, France
- Sorbonne Paris Cité, Paris Descartes University, 75015 Paris, France
| | - Eric Tartour
- Pediatric Nephrology, Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA) Reference Center, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
- PARCC, INSERM, Université Paris Cité, 75015 Paris, France
- Department of Immunology, Hôpital Européen Georges-Pompidou, AP-HP, CEDEX 15, 75908 Paris, France
| | - Maria Leite-de-Moraes
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Guillaume Lezmi
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie et Allergologie Pédiatriques, 75015 Paris, France
| | - Mickael Ménager
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, Université Paris Cité, Imagine Institute, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, 75015 Paris, France
| | - Marine Luka
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, Université Paris Cité, Imagine Institute, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, 75015 Paris, France
| | - Claude-Agnès Reynaud
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
| | - Jean-Claude Weill
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
| | - Laetitia Languille
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Marc Michel
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Pascal Chappert
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
- INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Thierry Mora
- Laboratoire de physique de l'École normale supérieure, CNRS, Paris Sciences et Lettres (PSL) University, Sorbonne Université, and Université de Paris, 75005 Paris, France
| | - Aleksandra M Walczak
- Laboratoire de physique de l'École normale supérieure, CNRS, Paris Sciences et Lettres (PSL) University, Sorbonne Université, and Université de Paris, 75005 Paris, France
| | - Marc Eloit
- Pathogen Discovery Laboratory, Institut Pasteur, Université Paris Cité, and Institut Pasteur, the WOAH Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Université Paris Cité, 75015 Paris, France
- Ecole Nationale Vétérinaire d'Alfort, University of Paris-Est, 94700 Maisons-Alfort, France
| | - Petra Bacher
- Institute of Immunology, Christian-Albrecht Universität zu Kiel and UKSH Schleswig-Holstein, 24105 Kiel, Germany
- Institute of Clinical Molecular Biology, Christian-Albrecht University of Kiel and UKSH Schleswig-Holstein, 24105 Kiel, Germany
| | - Alexander Scheffold
- Institute of Immunology, Christian-Albrecht Universität zu Kiel and UKSH Schleswig-Holstein, 24105 Kiel, Germany
| | - Matthieu Mahévas
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
- INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Isabelle Sermet-Gaudelus
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Reference Center for Rare Diseases: Cystic Fibrosis and Other Epithelial Respiratory Protein Misfolding Diseases, Hôpital Necker-Enfants Malades, AP-HP Centre Université Paris Cité, 75015 Paris, France
| | - Simon Fillatreau
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France
- Faculté de Médecine, Université Paris Cité, 75015 Paris, France
- Service Immunologie Biologique, AP-HP, Hôpital Universitaire Necker-Enfants Malades, F-75015 Paris, France
| |
Collapse
|
4
|
van der Lans SPA, Bardoel BW, Ruyken M, de Haas CJC, Baijens S, Muts RM, Scheepmaker LM, Aerts PC, van 't Wout MFL, Preiner J, Marijnissen RJ, Schuurman J, Beurskens FJ, Kerkman PF, Rooijakkers SHM. Agnostic B cell selection approach identifies antibodies against K. pneumoniae that synergistically drive complement activation. Nat Commun 2024; 15:8100. [PMID: 39285158 PMCID: PMC11405761 DOI: 10.1038/s41467-024-52372-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024] Open
Abstract
Antibody-dependent complement activation plays a key role in the natural human immune response to infections. Currently, the understanding of which antibody-antigen combinations drive a potent complement response on bacteria is limited. Here, we develop an antigen-agnostic approach to stain and single-cell sort human IgG memory B cells recognizing intact bacterial cells, keeping surface antigens in their natural context. With this method we successfully identified 29 antibodies against K. pneumoniae, a dominant cause of hospital-acquired infections with increasing antibiotic resistance. Combining genetic tools and functional analyses, we reveal that the capacity of antibodies to activate complement on K. pneumoniae critically depends on their antigenic target. Furthermore, we find that antibody combinations can synergistically activate complement on K. pneumoniae by strengthening each other's binding in an Fc-independent manner. Understanding the molecular basis of effective complement activation by antibody combinations to mimic a polyclonal response could accelerate the development of antibody-based therapies against problematic infections.
Collapse
Affiliation(s)
- Sjors P A van der Lans
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Bart W Bardoel
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Maartje Ruyken
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Carla J C de Haas
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Stan Baijens
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Remy M Muts
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Lisette M Scheepmaker
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Piet C Aerts
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marije F L van 't Wout
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | - Priscilla F Kerkman
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Suzan H M Rooijakkers
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
5
|
Kotaki R, Moriyama S, Oishi S, Onodera T, Adachi Y, Sasaki E, Ishino K, Morikawa M, Takei H, Takahashi H, Takano T, Nishiyama A, Yumoto K, Terahara K, Isogawa M, Matsumura T, Shinkai M, Takahashi Y. Repeated Omicron exposures redirect SARS-CoV-2-specific memory B cell evolution toward the latest variants. Sci Transl Med 2024; 16:eadp9927. [PMID: 39167666 DOI: 10.1126/scitranslmed.adp9927] [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: 04/22/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024]
Abstract
Immunological imprinting by ancestral SARS-CoV-2 strains is thought to impede the robust induction of Omicron-specific humoral responses by Omicron-based booster vaccines. Here, we analyzed the specificity and neutralization activity of memory B (Bmem) cells after repeated BA.5 exposure in individuals previously imprinted by ancestral strain-based mRNA vaccines. After a second BA.5 exposure, Bmem cells with BA.5 spike protein-skewed reactivity were promptly elicited, correlating with preexisting antibody titers. Clonal lineage analysis identified BA.5-skewed Bmem cells that had redirected their specificity from the ancestral strain to BA.5 through somatic hypermutations. Moreover, Bmem cells with redirected BA.5 specificity exhibited accelerated development compared with de novo Bmem cells derived from naïve repertoires. This redirected BA.5 specificity demonstrated greater resilience to viral point mutation and adaptation to recent Omicron variants HK.3 and JN.1, months after the second BA.5 exposure, suggesting that existing Bmem cells elicited by older vaccines can redirect their specificity toward newly evolving variants.
Collapse
Affiliation(s)
- Ryutaro Kotaki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Saya Moriyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Shintaro Oishi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Taishi Onodera
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Yu Adachi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Eita Sasaki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kota Ishino
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | | | | | | | - Tomohiro Takano
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Ayae Nishiyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kohei Yumoto
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kazutaka Terahara
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Masanori Isogawa
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Takayuki Matsumura
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | | | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Hokkaido 001-0021, Japan
| |
Collapse
|
6
|
Yang L, Zeng T, Li Y, Guo Q, Jiang D. Poor immune response to inactivated COVID-19 vaccine in patients with hypertension. Front Med (Lausanne) 2024; 11:1329607. [PMID: 38756945 PMCID: PMC11096495 DOI: 10.3389/fmed.2024.1329607] [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: 10/29/2023] [Accepted: 01/03/2024] [Indexed: 05/18/2024] Open
Abstract
Purpose The safety and efficacy of vaccination in people with hypertension (HTN) is important. There are currently a few data on the immunogenicity and safety of inactivated SARS-CoV-2 vaccinations in hypertension patients. Methods After receiving a two-dose immunization, 94 hypertension adult patients and 74 healthy controls (HCs) in this study, the evaluation included looking at antibodies (Abs) against receptor binding domain (RBD) IgG, SARS-CoV-2 neutralizing antibodies (NAbs), RBD-specific B cells, and memory B cells (MBCs). Results There was no discernible difference in the overall adverse events (AEs) over the course of 7 or 30 days between HTN patients and HCs. HTN patients had lower frequencies of RBD-specific memory B cells and the seropositivity rates and titers of Abs compared with HCs (all, p < 0.05). HTN patients with cardiovascular and cerebrovascular conditions (CCVD) have lower titers of CoV-2 NAb than in HCs. The titers of both Abs in HTN declined gradually over time. Conclusion Inactivated COVID-19 vaccinations were safe in hypertension patients; however humoral immune was limited, especially merged CCVD and declined gradually over time.
Collapse
Affiliation(s)
- Lei Yang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - TingTing Zeng
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yang Li
- Department of Respiratory Medicine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qiao Guo
- Department of General and Practice, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - DePeng Jiang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Ho VWT, Boon LH, Cui J, Juequn Z, Shunmuganathan B, Gupta R, Tan NYJ, Qian X, Purushotorman K, Fong S, Renia L, Ng LFP, Angeli V, Chen J, Kennedy BK, Ong CWM, Macary PA. Relative deficiency in interferon-γ-secreting CD4+ T cells is strongly associated with poorer COVID-19 vaccination responses in older adults. Aging Cell 2024; 23:e14099. [PMID: 38317404 PMCID: PMC11019126 DOI: 10.1111/acel.14099] [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: 08/02/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 02/07/2024] Open
Abstract
Although the two-dose mRNA vaccination regime provides protection against SARS-CoV-2, older adults have been shown to exhibit poorer vaccination responses. In addition, the role of vaccine-induced T-cell responses is not well characterised. We aim to assess the impact of age on immune responses after two doses of the BNT162b2 mRNA vaccine, focussing on antigen-specific T-cells. A prospective 3-month study was conducted on 15 young (median age 31 years, interquartile range (IQR) 25-35 years) and 14 older adults (median age 72 years, IQR 70-73 years). We assessed functional, neutralising antibody responses against SARS-CoV-2 variants using ACE-2 inhibition assays, and changes in B and T-cell subsets by high-dimensional flow cytometry. Antigen-specific T-cell responses were also quantified by intracellular cytokine staining and flow cytometry. Older adults had attenuated T-helper (Th) response to vaccination, which was associated with weaker antibody responses and decreased SARS-CoV-2 neutralisation. Antigen-specific interferon-γ (IFNγ)-secreting CD4+ T-cells to wild-type and Omicron antigens increased in young adults, which was strongly positively correlated with their neutralising antibody responses. Conversely, this relationship was negative in older adults. Hence, older adults' relative IFNγ-secreting CD4+ T cell deficiency might explain their poorer COVID-19 vaccination responses. Further exploration into the aetiology is needed and would be integral in developing novel vaccination strategies and improving infection outcomes in older adults.
Collapse
Affiliation(s)
- Vanda W. T. Ho
- Division of Geriatric Medicine, Department of MedicineNational University HospitalSingaporeSingapore
- Immunology Translational Research Program, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Infectious Diseases Translational Research Program, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Low Heng Boon
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Jianzhou Cui
- Immunology Translational Research Program, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- NUS Immunology Program, Life Sciences InstituteNational University of SingaporeSingaporeSingapore
- NUS‐Cambridge Immune Phenotyping Centre (NCIPC), Life Sciences InstituteNational University of SingaporeSingaporeSingapore
| | - Zhou Juequn
- Metabolic Core, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Bhuvaneshwari Shunmuganathan
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- NUS‐Cambridge Immune Phenotyping Centre (NCIPC), Life Sciences InstituteNational University of SingaporeSingaporeSingapore
- Antibody Engineering Programme, Life Sciences InstituteNational University of SingaporeSingaporeSingapore
| | - Rashi Gupta
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- NUS‐Cambridge Immune Phenotyping Centre (NCIPC), Life Sciences InstituteNational University of SingaporeSingaporeSingapore
| | - Nikki Y. J. Tan
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- NUS‐Cambridge Immune Phenotyping Centre (NCIPC), Life Sciences InstituteNational University of SingaporeSingaporeSingapore
| | - Xinlei Qian
- Antibody Engineering Programme, Life Sciences InstituteNational University of SingaporeSingaporeSingapore
| | - Kiren Purushotorman
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- NUS‐Cambridge Immune Phenotyping Centre (NCIPC), Life Sciences InstituteNational University of SingaporeSingaporeSingapore
- Antibody Engineering Programme, Life Sciences InstituteNational University of SingaporeSingaporeSingapore
| | - Siew‐Wai Fong
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Laurent Renia
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Lisa F. P. Ng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Veronique Angeli
- Immunology Translational Research Program, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Jinmiao Chen
- Immunology Translational Research Program, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Singapore Immunology Network (SIgN)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Brian K. Kennedy
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Biochemistry and Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Catherine W. M. Ong
- Infectious Diseases Translational Research Program, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingaporeSingapore
- Division of Infectious Diseases, Department of MedicineNational University HospitalSingaporeSingapore
| | - Paul A. Macary
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- NUS‐Cambridge Immune Phenotyping Centre (NCIPC), Life Sciences InstituteNational University of SingaporeSingaporeSingapore
- Antibody Engineering Programme, Life Sciences InstituteNational University of SingaporeSingaporeSingapore
| |
Collapse
|
8
|
Pastore G, Polvere J, Fiorino F, Lucchesi S, Montesi G, Rancan I, Zirpoli S, Lippi A, Durante M, Fabbiani M, Tumbarello M, Montagnani F, Medaglini D, Ciabattini A. Homologous or heterologous administration of mRNA or adenovirus-vectored vaccines show comparable immunogenicity and effectiveness against the SARS-CoV-2 Omicron variant. Expert Rev Vaccines 2024; 23:432-444. [PMID: 38517153 DOI: 10.1080/14760584.2024.2333952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/19/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Heterologous prime-boost schedules have been employed in SARS-CoV-2 vaccination, yet additional data on immunogenicity and effectiveness are still needed. RESEARCH DESIGN AND METHODS Here, we measured the immunogenicity and effectiveness in the real-world setting of the mRNA booster dose in 181 subjects who had completed primary vaccination with ChAdOx1, BNT162b2, or mRNA1273 vaccines (IMMUNO_COV study; protocol code 18,869). The spike-specific antibody and B cell responses were analyzed up to 6 months after boosting. RESULTS After an initial slower antibody response, the heterologous ChAdOx1/mRNA prime-boost formulation elicited spike-specific IgG titers comparable to homologous approaches, while spike-specific B cells showed a higher percentage of CD21-CD27- atypical cells compared to homologous mRNA vaccination. Mixed combinations of BNT162b2 and mRNA-1273 elicited an immune response comparable with homologous strategies. Non-significant differences in the Relative Risk of infection, calculated over a period of 18 months after boosting, were reported among homologous or heterologous vaccination groups, indicating a comparable relative vaccine effectiveness. CONCLUSIONS Our data endorse the heterologous booster vaccination with mRNA as a valuable alternative to homologous schedules. This approach can serve as a solution in instances of formulation shortages and contribute to enhancing vaccine strategies for potential epidemics or pandemics.
Collapse
Affiliation(s)
- Gabiria Pastore
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Jacopo Polvere
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Department of Medicine and Surgery, LUM University "Giuseppe Degennaro"; Casamassima, Bari, Italy
| | - Simone Lucchesi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Giorgio Montesi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Ilaria Rancan
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, Siena, Italy
| | - Sara Zirpoli
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Arianna Lippi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, Siena, Italy
| | - Miriam Durante
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | | - Mario Tumbarello
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, Siena, Italy
| | - Francesca Montagnani
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, Siena, Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
9
|
Zhang H, Liu Y, Liu Z. Nanomedicine approaches against SARS-CoV-2 and variants. J Control Release 2024; 365:101-111. [PMID: 37951476 DOI: 10.1016/j.jconrel.2023.11.004] [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: 07/26/2023] [Revised: 10/27/2023] [Accepted: 11/03/2023] [Indexed: 11/14/2023]
Abstract
The world is grappling with the ongoing crisis of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), a global pandemic that continues to have a detrimental impact on public health and economies worldwide. The virus's relentless mutation has led to more transmissible, immune-evasive strains, thereby escalating the incidence of reinfection. This underscores the urgent need for highly effective and safe countermeasures against SARS-CoV-2 and its evolving variants. In the current context, nanomedicine presents an innovative and promising alternative to mitigate the impacts of this pandemic wave. It does so by harnessing the structural and functional properties at a nanoscale in a straightforward and adaptable manner. This review emphasizes the most recent progress in the development of nanovaccines, nanodecoys, and nanodisinfectants to tackle SARS-CoV-2 and its variants. Notably, the insights gained and strategies implemented in managing the ongoing pandemic may also offer invaluable guidance for the development of potent nanomedicines to combat future pandemics.
Collapse
Affiliation(s)
- Han Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China.
| | - Yanbin Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China.
| |
Collapse
|
10
|
Hong Q, Bai Z, Tian D. SARS-CoV-2 booster immunization: stimulating de novo B-cell response and antibody generation. MedComm (Beijing) 2023; 4:e465. [PMID: 38116062 PMCID: PMC10728763 DOI: 10.1002/mco2.465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/26/2023] [Accepted: 11/30/2023] [Indexed: 12/21/2023] Open
Affiliation(s)
- Qiaonan Hong
- Department of Thoracic SurgeryWest China Hospital, Sichuan UniversityChengduChina
- Lung Transplant Research LaboratoryInstitute of Thoracic Oncology, West China Hospital, Sichuan UniversityChengduChina
| | - Ziyi Bai
- Department of Thoracic SurgeryWest China Hospital, Sichuan UniversityChengduChina
- Lung Transplant Research LaboratoryInstitute of Thoracic Oncology, West China Hospital, Sichuan UniversityChengduChina
| | - Dong Tian
- Department of Thoracic SurgeryWest China Hospital, Sichuan UniversityChengduChina
- Lung Transplant Research LaboratoryInstitute of Thoracic Oncology, West China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
11
|
Sokal A, Barba-Spaeth G, Hunault L, Fernández I, Broketa M, Meola A, Fourati S, Azzaoui I, Vandenberghe A, Lagouge-Roussey P, Broutin M, Roeser A, Bouvier-Alias M, Crickx E, Languille L, Fournier M, Michel M, Godeau B, Gallien S, Melica G, Nguyen Y, Canoui-Poitrine F, Pirenne F, Megret J, Pawlotsky JM, Fillatreau S, Reynaud CA, Weill JC, Rey FA, Bruhns P, Mahévas M, Chappert P. SARS-CoV-2 Omicron BA.1 breakthrough infection drives late remodeling of the memory B cell repertoire in vaccinated individuals. Immunity 2023; 56:2137-2151.e7. [PMID: 37543032 DOI: 10.1016/j.immuni.2023.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/12/2023] [Accepted: 07/06/2023] [Indexed: 08/07/2023]
Abstract
How infection by a viral variant showing antigenic drift impacts a preformed mature human memory B cell (MBC) repertoire remains an open question. Here, we studied the MBC response up to 6 months after SARS-CoV-2 Omicron BA.1 breakthrough infection in individuals previously vaccinated with three doses of the COVID-19 mRNA vaccine. Longitudinal analysis, using single-cell multi-omics and functional analysis of monoclonal antibodies from RBD-specific MBCs, revealed that a BA.1 breakthrough infection mostly recruited pre-existing cross-reactive MBCs with limited de novo response against BA.1-restricted epitopes. Reorganization of clonal hierarchy and new rounds of germinal center reactions, however, combined to maintain diversity and induce progressive maturation of the MBC repertoire against common Hu-1 and BA.1, but not BA.5-restricted, SARS-CoV-2 Spike RBD epitopes. Such remodeling was further associated with a marked improvement in overall neutralizing breadth and potency. These findings have fundamental implications for the design of future vaccination booster strategies.
Collapse
Affiliation(s)
- 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, Créteil, France; Service de Médecine interne, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris Cité, Clichy, France; Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Giovanna Barba-Spaeth
- Institut Pasteur, Université de Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| | - Lise Hunault
- Institut Pasteur, Université de Paris Cité, INSERM UMR1222, Unit of Antibodies in Therapy and Pathology, Paris, France; Sorbonne University, ED394, Paris, France; Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013 Paris, France
| | - Ignacio Fernández
- Institut Pasteur, Université de Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| | - Matteo Broketa
- Institut Pasteur, Université de Paris Cité, INSERM UMR1222, Unit of Antibodies in Therapy and Pathology, Paris, France; Sorbonne University, ED394, Paris, France
| | - Annalisa Meola
- Institut Pasteur, Université de Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| | - Slim Fourati
- Département de Virologie, Bactériologie, Hygiène et Mycologie-Parasitologie, Centre Hospitalier Universitaire Henri Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Créteil, France; INSERM U955, équipe 18. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Imane Azzaoui
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, 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, Créteil, France; Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Pauline Lagouge-Roussey
- 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, Créteil, France; Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Manon Broutin
- 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, Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Anais Roeser
- 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, Créteil, France; Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Magali Bouvier-Alias
- Département de Virologie, Bactériologie, Hygiène et Mycologie-Parasitologie, Centre Hospitalier Universitaire Henri Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Créteil, France; INSERM U955, équipe 18. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Etienne Crickx
- 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, Créteil, France; Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Laetitia Languille
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Morgane Fournier
- 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, Créteil, France; Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Marc Michel
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Bertrand Godeau
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Sébastien Gallien
- Service de Maladies Infectieuses, Centre Hospitalier Universitaire Henri Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Giovanna Melica
- Service de Maladies Infectieuses, Centre Hospitalier Universitaire Henri Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Yann Nguyen
- Service de Médecine Interne, Centre Hospitalier Universitaire Cochin, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Florence Canoui-Poitrine
- Département de Santé Publique, Unité de Recherche Clinique (URC), CEpiA (Clinical Epidemiology and Ageing), EA 7376- Institut Mondor de Recherche Biomédicale (IMRB), Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - France Pirenne
- INSERM U955, équipe 18. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France; Etablissement Français du Sang (EFS) Ile de France, Créteil, France
| | - Jérôme Megret
- Plateforme de Cytométrie en Flux, Structure Fédérative de Recherche Necker, INSERM US24-CNRS UMS3633, Paris, France
| | - Jean-Michel Pawlotsky
- Département de Virologie, Bactériologie, Hygiène et Mycologie-Parasitologie, Centre Hospitalier Universitaire Henri Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Créteil, France; INSERM U955, équipe 18. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Simon Fillatreau
- Institut Necker Enfants Malades (INEM), INSERM U1151/CNRS UMR 8253, Université de Paris, Paris, France
| | - Claude-Agnès Reynaud
- 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, Créteil, France
| | - Jean-Claude Weill
- 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, Créteil, France
| | - Félix A Rey
- Institut Pasteur, Université de Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| | - Pierre Bruhns
- Institut Pasteur, Université de Paris Cité, INSERM UMR1222, Unit of Antibodies in Therapy and Pathology, 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, Créteil, France; Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, 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, Créteil, France; INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France.
| |
Collapse
|
12
|
Guo S, Zheng Y, Gao Z, Duan M, Liu S, Du P, Xu X, Xu K, Zhao X, Chai Y, Wang P, Zhao Q, Gao GF, Dai L. Dosing interval regimen shapes potency and breadth of antibody repertoire after vaccination of SARS-CoV-2 RBD protein subunit vaccine. Cell Discov 2023; 9:79. [PMID: 37507370 PMCID: PMC10382582 DOI: 10.1038/s41421-023-00585-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023] Open
Abstract
Vaccination with different vaccines has been implemented globally to counter the continuous COVID-19 pandemic. However, the vaccine-elicited antibodies have reduced efficiency against the highly mutated Omicron sub-variants. Previously, we developed a protein subunit COVID-19 vaccine called ZF2001, based on the dimeric receptor-binding domain (RBD). This vaccine has been administered using different dosing intervals in real-world setting. Some individuals received three doses of ZF2001, with a one-month interval between each dose, due to urgent clinical requirements. Others had an extended dosing interval of up to five months between the second and third dose, a standard vaccination regimen for the protein subunit vaccine against hepatitis B. In this study, we profile B cell responses in individuals who received three doses of ZF2001, and compared those with long or short dosing intervals. We observed that the long-interval group exhibited higher and broader serologic antibody responses. These responses were associated with the increased size and evolution of vaccine-elicited B-cell receptor repertoires, characterized by the elevation of expanded clonotypes and somatic hypermutations. Both groups of individuals generated substantial amounts of broadly neutralizing antibodies (bnAbs) against various SARS-CoV-2 variants, including Omicron sub-variants such as XBB. These bnAbs target four antigenic sites within the RBD. To determine the vulnerable site of SARS-CoV-2, we employed cryo-electron microscopy to identify the epitopes of highly potent bnAbs that targeted two major sites. Our findings provide immunological insights into the B cell responses elicited by RBD-based vaccine, and suggest that a vaccination regimen of prolonging time interval should be used in practice.
Collapse
Affiliation(s)
- Shuxin Guo
- Faculty of Health Sciences, University of Macau, Macau SAR, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yuxuan Zheng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Zhengrong Gao
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Minrun Duan
- School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Sheng Liu
- Department of Biology, Cryo-EM Center, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Pan Du
- Vazyme Biotech, Nanjing, Jiangsu, China
| | - XiaoYu Xu
- Vazyme Biotech, Nanjing, Jiangsu, China
| | - Kun Xu
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Xin Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Yan Chai
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Peiyi Wang
- Department of Biology, Cryo-EM Center, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Qi Zhao
- MoE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - George F Gao
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China.
| | - Lianpan Dai
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
13
|
Zhang L, Jiang Y, He J, Chen J, Qi R, Yuan L, Shao T, Zhao H, Chen C, Chen Y, Wang X, Lei X, Gao Q, Zhuang C, Zhou M, Ma J, Liu W, Yang M, Fu R, Wu Y, Chen F, Xiong H, Nie M, Chen Y, Wu K, Fang M, Wang Y, Zheng Z, Huang S, Ge S, Cheng SC, Zhu H, Cheng T, Yuan Q, Wu T, Zhang J, Chen Y, Zhang T, Li C, Qi H, Guan Y, Xia N. Intranasal influenza-vectored COVID-19 vaccine restrains the SARS-CoV-2 inflammatory response in hamsters. Nat Commun 2023; 14:4117. [PMID: 37433761 DOI: 10.1038/s41467-023-39560-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 06/19/2023] [Indexed: 07/13/2023] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants and "anatomical escape" characteristics threaten the effectiveness of current coronavirus disease 2019 (COVID-19) vaccines. There is an urgent need to understand the immunological mechanism of broad-spectrum respiratory tract protection to guide broader vaccines development. Here we investigate immune responses induced by an NS1-deleted influenza virus vectored intranasal COVID-19 vaccine (dNS1-RBD) which provides broad-spectrum protection against SARS-CoV-2 variants in hamsters. Intranasal delivery of dNS1-RBD induces innate immunity, trained immunity and tissue-resident memory T cells covering the upper and lower respiratory tract. It restrains the inflammatory response by suppressing early phase viral load post SARS-CoV-2 challenge and attenuating pro-inflammatory cytokine (Il6, Il1b, and Ifng) levels, thereby reducing excess immune-induced tissue injury compared with the control group. By inducing local cellular immunity and trained immunity, intranasal delivery of NS1-deleted influenza virus vectored vaccine represents a broad-spectrum COVID-19 vaccine strategy to reduce disease burden.
Collapse
Affiliation(s)
- Liang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Yao Jiang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Jinhang He
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Junyu Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Ruoyao Qi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Lunzhi Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Tiange Shao
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Hui Zhao
- National Institute for Food and Drug Control, 102629, Beijing, China
| | - Congjie Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Yaode Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Xijing Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Xing Lei
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Qingxiang Gao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Chunlan Zhuang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Ming Zhou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Jian Ma
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Wei Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Man Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Rao Fu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Yangtao Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Feng Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Hualong Xiong
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Meifeng Nie
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Yiyi Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Kun Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Mujin Fang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Yingbin Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Zizheng Zheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Shoujie Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Shengxiang Ge
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Shih Chin Cheng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Huachen Zhu
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, 999077, China
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU), Shantou University, 515063, Shantou, China
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Quan Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Ting Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China.
| | - Jun Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China.
| | - Yixin Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China.
| | - Tianying Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China.
| | - Changgui Li
- National Institute for Food and Drug Control, 102629, Beijing, China.
| | - Hai Qi
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, 100084, Beijing, China.
| | - Yi Guan
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, 999077, China.
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU), Shantou University, 515063, Shantou, China.
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China.
| |
Collapse
|
14
|
Giron CC, Laaksonen A, Barroso da Silva FL. Differences between Omicron SARS-CoV-2 RBD and other variants in their ability to interact with cell receptors and monoclonal antibodies. J Biomol Struct Dyn 2023; 41:5707-5727. [PMID: 35815535 DOI: 10.1080/07391102.2022.2095305] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/23/2022] [Indexed: 12/23/2022]
Abstract
SARS-CoV-2 remains a health threat with the continuous emergence of new variants. This work aims to expand the knowledge about the SARS-CoV-2 receptor-binding domain (RBD) interactions with cell receptors and monoclonal antibodies (mAbs). By using constant-pH Monte Carlo simulations, the free energy of interactions between the RBD from different variants and several partners (Angiotensin-Converting Enzyme-2 (ACE2) polymorphisms and various mAbs) were predicted. Computed RBD-ACE2-binding affinities were higher for two ACE2 polymorphisms (rs142984500 and rs4646116) typically found in Europeans which indicates a genetic susceptibility. This is amplified for Omicron (BA.1) and its sublineages BA.2 and BA.3. The antibody landscape was computationally investigated with the largest set of mAbs so far in the literature. From the 32 studied binders, groups of mAbs were identified from weak to strong binding affinities (e.g. S2K146). These mAbs with strong binding capacity and especially their combination are amenable to experimentation and clinical trials because of their high predicted binding affinities and possible neutralization potential for current known virus mutations and a universal coronavirus.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Carolina Corrêa Giron
- Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
- Universidade Federal do Triângulo Mineiro, Hospital de Clínicas, Uberaba, MG, Brazil
| | - Aatto Laaksonen
- Department of Materials and Environmental Chemistry, Arrhenius Laboratory, Stockholm University, Stockholm, Sweden
- State Key Laboratory of Materials-Oriented and Chemical Engineering, Nanjing Tech University, Nanjing, PR China
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, Petru Poni Institute of Macromolecular Chemistry, Iasi, Romania
- Department of Engineering Sciences and Mathematics, Division of Energy Science, Luleå University of Technology, Luleå, Sweden
- Department of Chemical and Geological Sciences, University of Cagliari, Monserrato, Italy
| | - Fernando Luís Barroso da Silva
- Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
15
|
Ciabattini A, Pastore G, Lucchesi S, Montesi G, Costagli S, Polvere J, Fiorino F, Pettini E, Lippi A, Ancillotti L, Tumbarello M, Fabbiani M, Montagnani F, Medaglini D. Trajectory of Spike-Specific B Cells Elicited by Two Doses of BNT162b2 mRNA Vaccine. Cells 2023; 12:1706. [PMID: 37443740 PMCID: PMC10340653 DOI: 10.3390/cells12131706] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
The mRNA vaccines for SARS-CoV-2 have demonstrated efficacy and immunogenicity in the real-world setting. However, most of the research on vaccine immunogenicity has been centered on characterizing the antibody response, with limited exploration into the persistence of spike-specific memory B cells. Here we monitored the durability of the memory B cell response up to 9 months post-vaccination, and characterized the trajectory of spike-specific B cell phenotypes in healthy individuals who received two doses of the BNT162b2 vaccine. To profile the spike-specific B cell response, we applied the tSNE and Cytotree automated approaches. Spike-specific IgA+ and IgG+ plasmablasts and IgA+ activated cells were observed 7 days after the second dose and disappeared 3 months later, while subsets of spike-specific IgG+ resting memory B cells became predominant 9 months after vaccination, and they were capable of differentiating into spike-specific IgG secreting cells when restimulated in vitro. Other subsets of spike-specific B cells, such as IgM+ or unswitched IgM+IgD+ or IgG+ double negative/atypical cells, were also elicited by the BNT162b2 vaccine and persisted up to month 9. The analysis of circulating spike-specific IgG, IgA, and IgM was in line with the plasmablasts observed. The longitudinal analysis of the antigen-specific B cell response elicited by mRNA-based vaccines provides valuable insights into our understanding of the immunogenicity of this novel vaccine platform destined for future widespread use, and it can help in guiding future decisions and vaccination schedules.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Gabiria Pastore
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Simone Lucchesi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Giorgio Montesi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Simone Costagli
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Jacopo Polvere
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
- Department of Medicine and Surgery, LUM University “Giuseppe Degennaro”, 70010 Casamassima, Italy
| | - Elena Pettini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Arianna Lippi
- Infectious and Tropical Diseases Unit, Department of Medical Sciences, University Hospital of Siena, 53100 Siena, Italy; (A.L.); (L.A.); (M.T.); (M.F.); (F.M.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Leonardo Ancillotti
- Infectious and Tropical Diseases Unit, Department of Medical Sciences, University Hospital of Siena, 53100 Siena, Italy; (A.L.); (L.A.); (M.T.); (M.F.); (F.M.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Mario Tumbarello
- Infectious and Tropical Diseases Unit, Department of Medical Sciences, University Hospital of Siena, 53100 Siena, Italy; (A.L.); (L.A.); (M.T.); (M.F.); (F.M.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Massimiliano Fabbiani
- Infectious and Tropical Diseases Unit, Department of Medical Sciences, University Hospital of Siena, 53100 Siena, Italy; (A.L.); (L.A.); (M.T.); (M.F.); (F.M.)
| | - Francesca Montagnani
- Infectious and Tropical Diseases Unit, Department of Medical Sciences, University Hospital of Siena, 53100 Siena, Italy; (A.L.); (L.A.); (M.T.); (M.F.); (F.M.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| |
Collapse
|
16
|
Tamandjou Tchuem CR, Auvigne V, Vaux S, Montagnat C, Paireau J, Monnier Besnard S, Gabet A, Benhajkassen N, Le Strat Y, Parent Du Chatelet I, Levy-Bruhl D. Vaccine effectiveness and duration of protection of COVID-19 mRNA vaccines against Delta and Omicron BA.1 symptomatic and severe COVID-19 outcomes in adults aged 50 years and over in France. Vaccine 2023; 41:2280-2288. [PMID: 36870880 PMCID: PMC9968619 DOI: 10.1016/j.vaccine.2023.02.062] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/01/2023]
Abstract
The emergence of SARS-CoV-2 variants calls for continuous monitoring of vaccine effectiveness (VE). We estimated the absolute effectiveness of complete 2-dose primary vaccination and booster vaccination with COVID-19 mRNA vaccines, and the duration of protection against Delta and Omicron BA.1 symptomatic infection and severe outcomes. French residents aged ≥50 years, who presented with SARS-CoV-2-like symptoms and tested for SARS-CoV-2 between June 6, 2021 and February 10, 2022 were included. A test-negative study was conducted to estimate VE against symptomatic infection, using conditional logistic regression models. Cox proportional hazard regressions were performed to assess additional protection against severe COVID-19 outcomes (any hospitalization, and intensive care units [ICU] admission or in-hospital death). In total, 273732 cases and 735 919 controls were included. VE against symptomatic infection after 2-doses vaccination was 86% (95% CI: 75-92%) for Delta and 70% (58-79%) for Omicron, 7-30 days post vaccination. Protection waned over time, reaching 60% (57-63%) against Delta and 20% (16.-24%) for Omicron BA.1 > 120 days after vaccination. The booster dose fully restored protection against symtpomatic Delta infection (95% [81-99%]) but only partially against symptomatic Omicron BA.1 infection (63% [59-67%]). VE against Delta-related severe outcomes was above 95% with 2 doses, and persisted for at least four months. Protection against any Omicron BA.1-hospitalization was 92% (65%-99%) at 8-30 days, and 82% (67%-91%) > 120 days from the second dose. Against BA.1 ICU admission or in-patient death, VE stood at 98% (0-100%) at 8-30 days, and was 90% (40-99%) > 120 days from the second dose. Protection confered by mRNA vaccines against severe disease caused by either Delta or Omicron BA.1 appeared high and sustained over time. Protection against symptomatic diseases after 2 doses decreased rapidly, especially against Omicron BA.1. A booster dose restored high protection against Delta but only a partial one against Omicron BA.1.
Collapse
Affiliation(s)
- Cynthia Raissa Tamandjou Tchuem
- Direction des maladies infectieuses - Unité infections respiratoires et vaccination, Santé publique France, 12 rue du Val d'Osne, Saint-Maurice 94415, France.
| | - Vincent Auvigne
- Direction des maladies infectieuses - Unité infections respiratoires et vaccination, Santé publique France, 12 rue du Val d'Osne, Saint-Maurice 94415, France.
| | - Sophie Vaux
- Direction des maladies infectieuses - Unité infections respiratoires et vaccination, Santé publique France, 12 rue du Val d'Osne, Saint-Maurice 94415, France.
| | | | - Juliette Paireau
- Direction des maladies infectieuses - Unité infections respiratoires et vaccination, Santé publique France, 12 rue du Val d'Osne, Saint-Maurice 94415, France; Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, Université Paris Cité, CNRS, UMR 2000, Paris, France.
| | - Stéphanie Monnier Besnard
- Direction des maladies non transmissibles et traumatismes, Unité traumatismes et Avancée en âge, Santé publique France, 12 rue du Val d'Osne, Saint-Maurice 94415, France.
| | - Amélie Gabet
- Direction des maladies non transmissibles et traumatismes, Unité Cardio-métabolique et respiratoire, Santé publique France, 12 rue du Val d'Osne, Saint-Maurice 94415, France.
| | - Nabil Benhajkassen
- Direction des maladies infectieuses - Unité infections respiratoires et vaccination, Santé publique France, 12 rue du Val d'Osne, Saint-Maurice 94415, France.
| | - Yann Le Strat
- Direction Appui, Traitements et Analyses de données, Santé publique France, 12 rue du Val d'Osne, Saint-Maurice 94415, France.
| | - Isabelle Parent Du Chatelet
- Direction des maladies infectieuses - Unité infections respiratoires et vaccination, Santé publique France, 12 rue du Val d'Osne, Saint-Maurice 94415, France.
| | - Daniel Levy-Bruhl
- Direction des maladies infectieuses - Unité infections respiratoires et vaccination, Santé publique France, 12 rue du Val d'Osne, Saint-Maurice 94415, France.
| |
Collapse
|
17
|
Chauvin C, Levillayer L, Roumier M, Nielly H, Roth C, Karnam A, Bonam SR, Bourgarit A, Dubost C, Bousquet A, Le Burel S, Mestiri R, Sene D, Galland J, Vasse M, Groh M, Le Marchand M, Vassord-Dang C, Gautier JF, Pham-Thi N, Verny C, Pitard B, Planchais C, Mouquet H, Paul R, Simon-Loriere E, Bayry J, Gilardin L, Sakuntabhai A. Tocilizumab-treated convalescent COVID-19 patients retain the cross-neutralization potential against SARS-CoV-2 variants. iScience 2023; 26:106124. [PMID: 36776936 PMCID: PMC9894676 DOI: 10.1016/j.isci.2023.106124] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/10/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Although tocilizumab treatment in severe and critical coronavirus disease 2019 (COVID-19) patients has proven its efficacy at the clinical level, there is little evidence supporting the effect of short-term use of interleukin-6 receptor blocking therapy on the B cell sub-populations and the cross-neutralization of SARS-CoV-2 variants in convalescent COVID-19 patients. We performed immunological profiling of 69 tocilizumab-treated and non-treated convalescent COVID-19 patients in total. We observed that SARS-CoV-2-specific IgG1 titers depended on disease severity but not on tocilizumab treatment. The plasma of both treated and non-treated patients infected with the ancestral variant exhibit strong neutralizing activity against the ancestral virus and the Alpha, Beta, and Delta variants of SARS-CoV-2, whereas the Gamma and Omicron viruses were less sensitive to seroneutralization. Overall, we observed that, despite the clinical benefits of short-term tocilizumab therapy in modifying the cytokine storm associated with COVID-19 infections, there were no modifications in the robustness of B cell and IgG responses to Spike antigens.
Collapse
Affiliation(s)
- Camille Chauvin
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France
| | - Laurine Levillayer
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France
| | - Mathilde Roumier
- Service de Médecine Interne, Hôpital Foch, 92151 Suresnes, France
| | - Hubert Nielly
- Service de Médecine Interne, Hôpital d'Instruction des Armées Bégin, 94160 Saint Mandé, France
| | - Claude Roth
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France
| | - Anupama Karnam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, 75006, France
| | - Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, 75006, France
| | - Anne Bourgarit
- Hôpital Jean Verdier, HUPSSD, AP-HP, 93140 Bondy, France.,Sorbonne Paris-Nord University (Paris 13), 93000 Bobigny, France.,Inserm, UMR 1135 CIMI, 75013 Paris, France
| | - Clément Dubost
- Service de réanimation, Hôpital militaire Bégin, 94120 Saint Mandé, France.,Université Paris-Saclay, ENS Paris-Saclay, CNRS, Centre Borelli, 91190, Gif-sur-Yvette, France
| | - Aurore Bousquet
- Département des laboratoires, Hôpital militaire Bégin, 94120 Saint Mandé, France
| | - Sébastien Le Burel
- Service de Médecine Interne, Hôpital d'Instruction des Armées Bégin, 94160 Saint Mandé, France
| | - Raphaële Mestiri
- Service de Médecine Interne, Hôpital d'Instruction des Armées Bégin, 94160 Saint Mandé, France
| | - Damien Sene
- Département de médecine interne, Hôpital Lariboisière, Université de Paris (Diderot), AP-HP, 75010 Paris, France
| | - Joris Galland
- Département de médecine interne, Hôpital Lariboisière, Université de Paris (Diderot), AP-HP, 75010 Paris, France
| | - Marc Vasse
- Laboratoire de Biologie Médicale, Hôpital Foch, 92151 Suresnes, France.,UMRS-1176, Le Kremlin Bicêtre, France
| | - Matthieu Groh
- Service de Médecine Interne, Hôpital Foch, 92151 Suresnes, France
| | - Mathilde Le Marchand
- Department of Clinical Research and Innovation, Foch Hospital, 40 rue Worth, 92150 Suresnes, France
| | - Camille Vassord-Dang
- Department of Clinical Research and Innovation, Foch Hospital, 40 rue Worth, 92150 Suresnes, France
| | - Jean-François Gautier
- Departement of Diabetes and Endocrinology, Hôpital Lariboisière, APHP, and INSERM U1138 Paris, France.,Université de Paris, 75006 Paris, France
| | - Nhan Pham-Thi
- Unité de Neurophysiologie du Stress, Département des Neurosciences, Institut de Recherche Biomédicale des Armées (IRBA), BP 73 91223 Brétigny sur Orge Cedex, France
| | - Christiane Verny
- Unité de Neurophysiologie du Stress, Département des Neurosciences, Institut de Recherche Biomédicale des Armées (IRBA), BP 73 91223 Brétigny sur Orge Cedex, France
| | - Bruno Pitard
- Nantes Université, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT,UMR 1302, F-44000 Nantes, France
| | - Cyril Planchais
- Institut Pasteur, Université de Paris, Humoral Immunology Unit, Department of Immunology, 75015 Paris, France
| | - Hugo Mouquet
- Institut Pasteur, Université de Paris, Humoral Immunology Unit, Department of Immunology, 75015 Paris, France
| | - Richard Paul
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France
| | - Etienne Simon-Loriere
- Institut Pasteur, Université de Paris, G5 Evolutionary Genomics of RNA viruses, 75015 Paris, France
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, 75006, France.,Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678623, India
| | - Laurent Gilardin
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, 75006, France.,Sorbonne Paris-Nord University (Paris 13), 93000 Bobigny, France.,Service de médecine interne, Hôpital Jean Verdier, HUPSSD, AP-HP, 93140 Bondy, France
| | - Anavaj Sakuntabhai
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France.,International Vaccine Design Center (vDesC), The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, Japan
| |
Collapse
|
18
|
Beukenhorst AL, Koch CM, Hadjichrysanthou C, Alter G, de Wolf F, Anderson RM, Goudsmit J. SARS-CoV-2 elicits non-sterilizing immunity and evades vaccine-induced immunity: implications for future vaccination strategies. Eur J Epidemiol 2023; 38:237-242. [PMID: 36738380 PMCID: PMC9898703 DOI: 10.1007/s10654-023-00965-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/07/2023] [Indexed: 02/05/2023]
Abstract
Neither vaccination nor natural infection result in long-lasting protection against SARS-COV-2 infection and transmission, but both reduce the risk of severe COVID-19. To generate insights into optimal vaccination strategies for prevention of severe COVID-19 in the population, we extended a Susceptible-Exposed-Infectious-Removed (SEIR) mathematical model to compare the impact of vaccines that are highly protective against severe COVID-19 but not against infection and transmission, with those that block SARS-CoV-2 infection. Our analysis shows that vaccination strategies focusing on the prevention of severe COVID-19 are more effective than those focusing on creating of herd immunity. Key uncertainties that would affect the choice of vaccination strategies are: (1) the duration of protection against severe disease, (2) the protection against severe disease from variants that escape vaccine-induced immunity, (3) the incidence of long-COVID and level of protection provided by the vaccine, and (4) the rate of serious adverse events following vaccination, stratified by demographic variables.
Collapse
Affiliation(s)
- Anna L Beukenhorst
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Centre for Epidemiology Versus Arthritis, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
- Leyden Laboratories BV, Amsterdam, The Netherlands.
| | | | | | - Galit Alter
- Ragon Institute of MGH MIT and Harvard, Cambridge, MA, USA
| | - Frank de Wolf
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
| | - Roy M Anderson
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
| | - Jaap Goudsmit
- Leyden Laboratories BV, Amsterdam, The Netherlands
- Departments of Epidemiology, Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
19
|
Yu H, Liu B, Zhang Y, Gao X, Wang Q, Xiang H, Peng X, Xie C, Wang Y, Hu P, Shi J, Shi Q, Zheng P, Feng C, Tang G, Liu X, Guo L, Lin X, Li J, Liu C, Huang Y, Yang N, Chen Q, Li Z, Su M, Yan Q, Pei R, Chen X, Liu L, Hu F, Liang D, Ke B, Ke C, Li F, He J, Wang M, Chen L, Xiong X, Tang X. Somatically hypermutated antibodies isolated from SARS-CoV-2 Delta infected patients cross-neutralize heterologous variants. Nat Commun 2023; 14:1058. [PMID: 36828833 PMCID: PMC9951844 DOI: 10.1038/s41467-023-36761-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 02/10/2023] [Indexed: 02/26/2023] Open
Abstract
SARS-CoV-2 Omicron variants feature highly mutated spike proteins with extraordinary abilities in evading antibodies isolated earlier in the pandemic. Investigation of memory B cells from patients primarily with breakthrough infections with the Delta variant enables isolation of a number of neutralizing antibodies cross-reactive to heterologous variants of concern (VOCs) including Omicron variants (BA.1-BA.4). Structural studies identify altered complementarity determining region (CDR) amino acids and highly unusual heavy chain CDR2 insertions respectively in two representative cross-neutralizing antibodies-YB9-258 and YB13-292. These features are putatively introduced by somatic hypermutation and they are heavily involved in epitope recognition to broaden neutralization breadth. Previously, insertions/deletions were rarely reported for antiviral antibodies except for those induced by HIV-1 chronic infections. These data provide molecular mechanisms for cross-neutralization of heterologous SARS-CoV-2 variants by antibodies isolated from Delta variant infected patients with implications for future vaccination strategy.
Collapse
Affiliation(s)
- Haisheng Yu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Banghui Liu
- The State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, the Chinese Academy of Sciences, Guangzhou, China
| | - Yudi Zhang
- The State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, the Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xijie Gao
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, China
| | - Qian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haitao Xiang
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Xiaofang Peng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Caixia Xie
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Yaping Wang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Peiyu Hu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, China
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Jingrong Shi
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Quan Shi
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Pingqian Zheng
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, China
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Chengqian Feng
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Guofang Tang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaopan Liu
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Liliangzi Guo
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiumei Lin
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Jiaojiao Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chuanyu Liu
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Yaling Huang
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Naibo Yang
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Qiuluan Chen
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, China
| | - Zimu Li
- The State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, the Chinese Academy of Sciences, Guangzhou, China
| | - Mengzhen Su
- The State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, the Chinese Academy of Sciences, Guangzhou, China
- University of Science and Technology of China, Hefei, Anhui, China
| | - Qihong Yan
- The State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, the Chinese Academy of Sciences, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Rongjuan Pei
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xinwen Chen
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, China
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Longqi Liu
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Fengyu Hu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Dan Liang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Bixia Ke
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Changwen Ke
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China.
| | - Feng Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Jun He
- The State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, the Chinese Academy of Sciences, Guangzhou, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, China.
| | - Meiniang Wang
- BGI-Shenzhen, Shenzhen, China.
- China National GeneBank, BGI-Shenzhen, Shenzhen, China.
| | - Ling Chen
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China.
- The State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, the Chinese Academy of Sciences, Guangzhou, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, China.
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, China.
| | - Xiaoli Xiong
- The State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, the Chinese Academy of Sciences, Guangzhou, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, China.
| | - Xiaoping Tang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China.
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, China.
| |
Collapse
|
20
|
Zappa M, Verdecchia P, Angeli F. The new phase of pandemic: Are BA.2.75 and BQ.1 competitive variants? An in silico evaluation. Eur J Intern Med 2023; 108:106-107. [PMID: 36357251 DOI: 10.1016/j.ejim.2022.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Martina Zappa
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Paolo Verdecchia
- Fondazione Umbra Cuore e Ipertensione-ONLUS and Division of Cardiology, Hospital S. Maria della Misericordia, Perugia, Italy
| | - Fabio Angeli
- Department of Medicine and Surgery, University of Insubria, Varese, Italy; Istituti Clinici Scientifici Maugeri, IRCCS Tradate, Italy.
| |
Collapse
|
21
|
Kotaki R, Moriyama S, Takahashi Y. Humoral immunity for durable control of SARS-CoV-2 and its variants. Inflamm Regen 2023; 43:4. [PMID: 36631890 PMCID: PMC9834039 DOI: 10.1186/s41232-023-00255-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is ongoing because of the repeated emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants, highlighting the importance of developing vaccines for variants that may continue to emerge. In the present review, we discuss humoral immune responses against SARS-CoV-2 with a focus on the antibody breadth to the variants. Recent studies have revealed that the temporal maturation of humoral immunity improves the antibody potency and breadth to the variants after infection or vaccination. Repeated vaccination or infection further accelerates the expansion of the antibody breadth. Memory B cells play a central role in this phenomenon, as the reactivity of the B-cell antigen receptor (BCR) on memory B cells is a key determinant of the antibody potency and breadth recalled upon vaccination or infection. The evolution of memory B cells remarkably improves the reactivity of BCR to antigenically distinct Omicron variants, to which the host has never been exposed. Thus, the evolution of memory B cells toward the variants constitutes an immunological basis for the durable and broad control of SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Ryutaro Kotaki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Saya Moriyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan.
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan.
| |
Collapse
|
22
|
Sokal A, Bastard P, Chappert P, Barba-Spaeth G, Fourati S, Vanderberghe A, Lagouge-Roussey P, Meyts I, Gervais A, Bouvier-Alias M, Azzaoui I, Fernández I, de la Selle A, Zhang Q, Bizien L, Pellier I, Linglart A, Rothenbuhler A, Marcoux E, Anxionnat R, Cheikh N, Léger J, Amador-Borrero B, Fouyssac F, Menut V, Goffard JC, Storey C, Demily C, Mallebranche C, Troya J, Pujol A, Zins M, Tiberghien P, Gray PE, McNaughton P, Sullivan A, Peake J, Levy R, Languille L, Rodiguez-Gallego C, Boisson B, Gallien S, Neven B, Michel M, Godeau B, Abel L, Rey FA, Weill JC, Reynaud CA, Tangye SG, Casanova JL, Mahévas M. Human type I IFN deficiency does not impair B cell response to SARS-CoV-2 mRNA vaccination. J Exp Med 2023; 220:213666. [PMID: 36342455 DOI: 10.1084/jem.20220258] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 08/02/2022] [Accepted: 10/05/2022] [Indexed: 11/09/2022] Open
Abstract
Inborn and acquired deficits of type I interferon (IFN) immunity predispose to life-threatening COVID-19 pneumonia. We longitudinally profiled the B cell response to mRNA vaccination in SARS-CoV-2 naive patients with inherited TLR7, IRF7, or IFNAR1 deficiency, as well as young patients with autoantibodies neutralizing type I IFNs due to autoimmune polyendocrine syndrome type-1 (APS-1) and older individuals with age-associated autoantibodies to type I IFNs. The receptor-binding domain spike protein (RBD)-specific memory B cell response in all patients was quantitatively and qualitatively similar to healthy donors. Sustained germinal center responses led to accumulation of somatic hypermutations in immunoglobulin heavy chain genes. The amplitude and duration of, and viral neutralization by, RBD-specific IgG serological response were also largely unaffected by TLR7, IRF7, or IFNAR1 deficiencies up to 7 mo after vaccination in all patients. These results suggest that induction of type I IFN is not required for efficient generation of a humoral response against SARS-CoV-2 by mRNA vaccines.
Collapse
Affiliation(s)
- Aurélien Sokal
- Necker Enfants Malades Institute, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team Auto-Immune and Immune B cell, University Paris Cité, University Paris-Est-Créteil, Créteil, France
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France.,Imagine Institute, University Paris Cité, Paris, France.,Department of Pediatrics, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris, Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Pascal Chappert
- Necker Enfants Malades Institute, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team Auto-Immune and Immune B cell, University Paris Cité, University Paris-Est-Créteil, Créteil, France.,INSERM U955, team 2. Mondor Biomedical Research Institute, Paris-Est Créteil University, Créteil, France
| | - Giovanna Barba-Spaeth
- Institut Pasteur, University Paris Cité, CNRS UMR 3569, Structural Virology Unit, Paris, France
| | - Slim Fourati
- Virology, Bacteriology, Hygiene and Mycology-Parasitology, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France.,INSERM U955, team 18. Mondor Biomedical Research Institute, Paris-Est Créteil University, Créteil, France
| | - Alexis Vanderberghe
- INSERM U955, team 2. Mondor Biomedical Research Institute, Paris-Est Créteil University, Créteil, France.,Departement of Internal Medicine, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Paris-Est Créteil University, Créteil, France
| | - Pauline Lagouge-Roussey
- INSERM U955, team 2. Mondor Biomedical Research Institute, Paris-Est Créteil University, Créteil, France.,Departement of Internal Medicine, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Paris-Est Créteil University, Créteil, France
| | - Isabelle Meyts
- Department of Immunology and Microbiology, Laboratory for Inborn Errors of Immunity, Department of Pediatrics, University Hospitals Leuven and Katholieke Universiteit Leuven, Leuven, Belgium
| | - Adrian Gervais
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France.,Imagine Institute, University Paris Cité, Paris, France
| | - Magali Bouvier-Alias
- Virology, Bacteriology, Hygiene and Mycology-Parasitology, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France.,INSERM U955, team 18. Mondor Biomedical Research Institute, Paris-Est Créteil University, Créteil, France
| | - Imane Azzaoui
- INSERM U955, team 2. Mondor Biomedical Research Institute, Paris-Est Créteil University, Créteil, France.,Departement of Internal Medicine, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Paris-Est Créteil University, Créteil, France
| | - Ignacio Fernández
- Institut Pasteur, University Paris Cité, CNRS UMR 3569, Structural Virology Unit, Paris, France
| | - Andréa de la Selle
- Necker Enfants Malades Institute, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team Auto-Immune and Immune B cell, University Paris Cité, University Paris-Est-Créteil, Créteil, France
| | - Qian Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France.,Imagine Institute, University Paris Cité, Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Lucy Bizien
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France.,Imagine Institute, University Paris Cité, Paris, France
| | - Isabelle Pellier
- Pediatric Immuno-hemato-oncology Unit, Centre Hospitalier Universitaire Angers, Angers, France.,University Angers, Nantes university, Centre Hospitalier Universitaire Angers, INSERM, CRCI2NA, SFR ICAT, Angers, France
| | - Agnès Linglart
- Departement of Pediatric Endocrinology, Bicêtre University Hospital, Assistance Publique-Hôpitaux de Paris, Paris Saclay University, Le Kremlin-Bicêtre, France
| | - Anya Rothenbuhler
- Departement of Pediatric Endocrinology, Bicêtre University Hospital, Assistance Publique-Hôpitaux de Paris, Paris Saclay University, Le Kremlin-Bicêtre, France
| | - Estelle Marcoux
- Department of Pediatrics, Nord Franche Comté Hospital, Trévenans, France
| | | | - Nathalie Cheikh
- Department of Pediatrics, Besançon Hospital, Besançon, France
| | - Juliane Léger
- Department of Pediatric Endocrinology and INSERM NeuroDiderot, Referral Centre for Endocrine, Growth and Development diseases, Assistance Publique-Hôpitaux de Paris Nord, University of Paris, Paris, France
| | - Blanca Amador-Borrero
- Department of Internal Medicine, Lariboisière University Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris, Paris, France
| | - Fanny Fouyssac
- Department of Pediatric Hemato-oncology, Childrens Hospital, Nancy University Hospital, Nancy, France
| | - Vanessa Menut
- Department of Pediatrics, Mother-Child Hospital, Nantes, France
| | - Jean-Christophe Goffard
- Department of Internal Medicine, Université Libre de Bruxelles-Hôpitaux Universitaire de Bruxelles, Erasme Hospital, Bruxelles, Belgique
| | - Caroline Storey
- Departement of Pediatric Endocrinology, Robert Debré Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Caroline Demily
- GénoPsy Referral Center, Centre de Référence de Maladies Rares Rare Disease with Psychiatric Epression, Le Vinatier Hospital, Bron, France
| | - Coralie Mallebranche
- Pediatric Immuno-hemato-oncology Unit, Centre Hospitalier Universitaire Angers, Angers, France.,University Angers, Nantes university, Centre Hospitalier Universitaire Angers, INSERM, CRCI2NA, SFR ICAT, Angers, France
| | - Jesus Troya
- Department of Internal Medicine, Infanta Leonor University Hospital, Madrid, Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge-Hospital Duran i Reynals, Centro de Investigación Biomédica en Red de Enfermededas Raras U759, and Catalan Institution of Research and Advanced Studies, Barcelona, Spain
| | - Marie Zins
- University of Paris, University of Paris-Saclay, Université de Versailles Saint-Quentin-en-Yvelines, INSERM UMS11, Villejuif, France
| | - Pierre Tiberghien
- French Blood Agency, La Plaine Saint-Denis, France.,UMR1098 RIGHT, INSERM, French Blood Agency, Franche-Comté University, Besançon, France
| | - Paul E Gray
- Department of Immunology and Infectious Diseases, Sydney Children's Hospital, Randwick, New South Wales, Australia.,School of Women's and Children's Health, University of New South Wales, Sydney, Australia.,Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia
| | - Peter McNaughton
- Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia.,Queensland Paediatric Immunology and Allergy Service, Children's Health Queensland, Brisbane, Australia
| | - Anna Sullivan
- Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia.,Queensland Paediatric Immunology and Allergy Service, Children's Health Queensland, Brisbane, Australia
| | - Jane Peake
- Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia.,Queensland Paediatric Immunology and Allergy Service, Children's Health Queensland, Brisbane, Australia.,University of Queensland, Brisbane, Australia
| | - Romain Levy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France.,Imagine Institute, University Paris Cité, Paris, France.,Department of Pediatric Immuno-Haematology and Rheumatology, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Laetitia Languille
- Departement of Internal Medicine, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Paris-Est Créteil University, Créteil, France
| | - Carlos Rodiguez-Gallego
- Department of Immunology, Hospital Universitario de G.C. Dr. Negrín, Canarian Health System, Las Palmas de Gran Canaria, Spain.,University Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Bertrand Boisson
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France.,Imagine Institute, University Paris Cité, Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Sébastien Gallien
- Department of Infectious Diseases, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Paris-Est Créteil University, Créteil, France
| | - Bénédicte Neven
- Department of Pediatric Immuno-Haematology and Rheumatology, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marc Michel
- Departement of Internal Medicine, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Paris-Est Créteil University, Créteil, France
| | - Bertrand Godeau
- Departement of Internal Medicine, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Paris-Est Créteil University, Créteil, France
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France.,Imagine Institute, University Paris Cité, Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Felix A Rey
- Institut Pasteur, University Paris Cité, CNRS UMR 3569, Structural Virology Unit, Paris, France
| | - Jean-Claude Weill
- Necker Enfants Malades Institute, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team Auto-Immune and Immune B cell, University Paris Cité, University Paris-Est-Créteil, Créteil, France
| | - Claude-Agnès Reynaud
- Necker Enfants Malades Institute, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team Auto-Immune and Immune B cell, University Paris Cité, University Paris-Est-Créteil, Créteil, France
| | - Stuart G Tangye
- Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia.,Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia.,St Vincent's Clinical School, Faculty of Medicine & Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France.,Imagine Institute, University Paris Cité, Paris, France.,Department of Pediatrics, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris, Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY.,Howard Hughes Medical Institute, New York, NY
| | - Matthieu Mahévas
- Necker Enfants Malades Institute, INSERM U1151/CNRS UMR 8253, Action thématique incitative sur programme-Avenir Team Auto-Immune and Immune B cell, University Paris Cité, University Paris-Est-Créteil, Créteil, France.,INSERM U955, team 2. Mondor Biomedical Research Institute, Paris-Est Créteil University, Créteil, France.,Departement of Internal Medicine, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Paris-Est Créteil University, Créteil, France
| |
Collapse
|
23
|
Ruhl L, Kühne JF, Beushausen K, Keil J, Christoph S, Sauer J, Falk CS. Third SARS-CoV-2 vaccination and breakthrough infections enhance humoral and cellular immunity against variants of concern. Front Immunol 2023; 14:1120010. [PMID: 37033958 PMCID: PMC10073596 DOI: 10.3389/fimmu.2023.1120010] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction SARS-CoV-2 vaccination is the leading strategy to prevent severe courses after SARS-CoV-2 infection. In our study, we analyzed humoral and cellular immune responses in detail to three consecutive homologous or heterologous SARS-CoV-2 vaccinations and breakthrough infections. Methods Peripheral blood samples of n=20 individuals were analyzed in the time course of three SARS-CoV-2 vaccinations and/or breakthrough infection. S1-, RBD-, S2- and N-specific IgG antibodies were quantified using Luminex-based multiplex assays and electrochemiluminescence multiplex assays for surrogate neutralization in plasma. Changes in cellular immune components were determined via flow cytometry of whole blood samples. Results All individuals (n=20) responded to vaccination with increasing S1-/RBD-/S2-specific IgG levels, whereas specific plasma IgA displayed individual variability. The third dose increased antibody inhibitory capacity (AIC) against immune-escape variants Beta and Omicron BA.1 independently of age. The mRNA-primed vaccination induced IgG and IgA immunity more efficiently, whereas vector-primed individuals displayed higher levels of memory T and B cells. Vaccinees showed SARS-CoV-2-specific T cell responses, which were further improved and specified after Omicron breakthrough infections in parallel to the appearance of new variant-specific antibodies. Discussion In conclusion, the third vaccination was essential to increase IgG levels, mandatory to boost AIC against immune-escape variants, and induced SARS-CoV-2-specific T cells. Breakthrough infection with Omicron generates additional spike specificities covering all known variants.
Collapse
Affiliation(s)
- Louisa Ruhl
- Hannover Medical School, Institute of Transplant Immunology, Hannover, Germany
| | - Jenny F. Kühne
- Hannover Medical School, Institute of Transplant Immunology, Hannover, Germany
| | - Kerstin Beushausen
- Hannover Medical School, Institute of Transplant Immunology, Hannover, Germany
| | - Jana Keil
- Hannover Medical School, Institute of Transplant Immunology, Hannover, Germany
| | - Stella Christoph
- Hannover Medical School, Institute of Transplant Immunology, Hannover, Germany
| | - Jasper Sauer
- Hannover Medical School, Institute of Transplant Immunology, Hannover, Germany
| | - Christine S. Falk
- Hannover Medical School, Institute of Transplant Immunology, Hannover, Germany
- BREATH Site, German Center for Lung Research (DZL), Hannover, Germany
- TTU-IICH, German Center for Infection Research (DZIF), Hannover-Braunschweig, Germany
- *Correspondence: Christine S. Falk,
| |
Collapse
|
24
|
Frans G, Dillaerts D, Dehaemers T, Van Elslande J, De Leeuw J, Boon L, Maes W, Callewaert N, Calcoen B, Ancheva L, Cockx M, Geukens N, Arat K, Derua R, Vermeersch P, Carpentier SC, Bossuyt X. Complementarity determining regions in SARS-CoV-2 hybrid immunity. Front Immunol 2023; 14:1050037. [PMID: 36895570 PMCID: PMC9990870 DOI: 10.3389/fimmu.2023.1050037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/02/2023] [Indexed: 02/23/2023] Open
Abstract
Pre-vaccination SARS-CoV-2 infection can boost protection elicited by COVID-19 vaccination and post-vaccination breakthrough SARS-CoV-2 infection can boost existing immunity conferred by COVID-19 vaccination. Such 'hybrid immunity' is effective against SARS-CoV-2 variants. In order to understand 'hybrid immunity' at the molecular level we studied the complementarity determining regions (CDR) of anti-RBD (receptor binding domain) antibodies isolated from individuals with 'hybrid immunity' as well as from 'naive' (not SARS-CoV-2 infected) vaccinated individuals. CDR analysis was done by liquid chromatography/mass spectrometry-mass spectrometry. Principal component analysis and partial least square differential analysis showed that COVID-19 vaccinated people share CDR profiles and that pre-vaccination SARS-CoV-2 infection or breakthrough infection further shape the CDR profile, with a CDR profile in hybrid immunity that clustered away from the CDR profile in vaccinated people without infection. Thus, our results show a CDR profile in hybrid immunity that is distinct from the vaccination-induced CDR profile.
Collapse
Affiliation(s)
- Glynis Frans
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Doreen Dillaerts
- Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Tom Dehaemers
- Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jan Van Elslande
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Jonas De Leeuw
- Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Lise Boon
- Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Wim Maes
- PharmAbs: The KU Leuven Antibody Center, KU Leuven, Leuven, Belgium
| | - Nico Callewaert
- Clinical Laboratory, AZ Groeninge Hospital, Kortrijk, Belgium
| | - Bas Calcoen
- Laboratory for Thrombosis Research, KU Leuven Kulak Kortrijk, Kortrijk, Belgium
| | - Lina Ancheva
- Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Maaike Cockx
- Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Nick Geukens
- PharmAbs: The KU Leuven Antibody Center, KU Leuven, Leuven, Belgium
| | | | - Rita Derua
- SyBioMa, KU Leuven, Leuven, Belgium.,Department of Molecular and Cellular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Leuven, Belgium
| | - Pieter Vermeersch
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium.,Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | | | - Xavier Bossuyt
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium.,Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Titers of antibodies against ancestral SARS-CoV-2 correlate with levels of neutralizing antibodies to multiple variants. NPJ Vaccines 2022; 7:174. [PMID: 36585405 PMCID: PMC9801350 DOI: 10.1038/s41541-022-00586-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/24/2022] [Indexed: 12/31/2022] Open
Abstract
Diagnostic assays currently used to monitor the efficacy of COVID-19 vaccines measure levels of antibodies to the receptor-binding domain of ancestral SARS-CoV-2 (RBDwt). However, the predictive value for protection against new variants of concern (VOCs) has not been firmly established. Here, we used bead-based arrays and flow cytometry to measure binding of antibodies to spike proteins and receptor-binding domains (RBDs) from VOCs in 12,000 serum samples. Effects of sera on RBD-ACE2 interactions were measured as a proxy for neutralizing antibodies. The samples were obtained from healthy individuals or patients on immunosuppressive therapy who had received two to four doses of COVID-19 vaccines and from COVID-19 convalescents. The results show that anti-RBDwt titers correlate with the levels of binding- and neutralizing antibodies against the Alpha, Beta, Gamma, Delta, Epsilon and Omicron variants. The benefit of multiplexed analysis lies in the ability to measure a wide range of anti-RBD titers using a single dilution of serum for each assay. The reactivity patterns also yield an internal reference for neutralizing activity and binding antibody units per milliliter (BAU/ml). Results obtained with sera from vaccinated healthy individuals and patients confirmed and extended results from previous studies on time-dependent waning of antibody levels and effects of immunosuppressive agents. We conclude that anti-RBDwt titers correlate with levels of neutralizing antibodies against VOCs and propose that our method may be implemented to enhance the precision and throughput of immunomonitoring.
Collapse
|
26
|
Cinicola BL, Piano Mortari E, Zicari AM, Agrati C, Bordoni V, Albano C, Fedele G, Schiavoni I, Leone P, Fiore S, Capponi M, Conti MG, Petrarca L, Stefanelli P, Spalice A, Midulla F, Palamara AT, Quinti I, Locatelli F, Carsetti R. The BNT162b2 vaccine induces humoral and cellular immune memory to SARS-CoV-2 Wuhan strain and the Omicron variant in children 5 to 11 years of age. Front Immunol 2022; 13:1094727. [PMID: 36591287 PMCID: PMC9797965 DOI: 10.3389/fimmu.2022.1094727] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
SARS-CoV-2 mRNA vaccines prevent severe COVID-19 by generating immune memory, comprising specific antibodies and memory B and T cells. Although children are at low risk of severe COVID-19, the spreading of highly transmissible variants has led to increasing in COVID-19 cases and hospitalizations also in the youngest, but vaccine coverage remains low. Immunogenicity to mRNA vaccines has not been extensively studied in children 5 to 11 years old. In particular, cellular immunity to the wild-type strain (Wuhan) and the cross-reactive response to the Omicron variant of concern has not been investigated. We assessed the humoral and cellular immune response to the SARS-CoV-2 BNT162b2 vaccine in 27 healthy children. We demonstrated that vaccination induced a potent humoral and cellular immune response in all vaccinees. By using spike-specific memory B cells as a measurable imprint of a previous infection, we found that 50% of the children had signs of a past, undiagnosed infection before vaccination. Children with pre-existent immune memory generated significantly increased levels of specific antibodies, and memory T and B cells, directed against not only the wild type virus but also the omicron variant.
Collapse
Affiliation(s)
- Bianca Laura Cinicola
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy,Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - E Piano Mortari
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy,B cell unit, Immunology Research Area, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Anna Maria Zicari
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Chiara Agrati
- Department of Pediatric Hematology and Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Veronica Bordoni
- Department of Pediatric Hematology and Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Christian Albano
- B cell unit, Immunology Research Area, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Giorgio Fedele
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Ilaria Schiavoni
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Pasqualina Leone
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Stefano Fiore
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Martina Capponi
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Maria Giulia Conti
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Laura Petrarca
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Paola Stefanelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Alberto Spalice
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Fabio Midulla
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | | | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy,Catholic University of the Sacred Hearth, Rome, Italy
| | - Rita Carsetti
- B cell unit, Immunology Research Area, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy,*Correspondence: Rita Carsetti,
| |
Collapse
|
27
|
Kardava L, Buckner CM, Moir S. B-Cell Responses to Sars-Cov-2 mRNA Vaccines. Pathog Immun 2022; 7:93-119. [PMID: 36655200 PMCID: PMC9836209 DOI: 10.20411/pai.v7i2.550] [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: 10/05/2022] [Accepted: 10/23/2022] [Indexed: 12/14/2022] Open
Abstract
Most vaccines against viral pathogens protect through the acquisition of immunological memory from long-lived plasma cells that produce antibodies and memory B cells that can rapidly respond upon an encounter with the pathogen or its variants. The COVID-19 pandemic and rapid deployment of effective vaccines have provided an unprecedented opportunity to study the immune response to a new yet rapidly evolving pathogen. Here we review the scientific literature and our efforts to understand antibody and B-cell responses to SARS-CoV-2 vaccines, the effect of SARSCoV-2 infection on both primary and secondary immune responses, and how repeated exposures may impact outcomes.
Collapse
Affiliation(s)
- Lela Kardava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD
| | - Clarisa M. Buckner
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD
| |
Collapse
|
28
|
Xiang T, Wang J, Zheng X. The humoral and cellular immune evasion of SARS-CoV-2 Omicron and sub-lineages. Virol Sin 2022; 37:786-795. [PMID: 36427646 PMCID: PMC9681989 DOI: 10.1016/j.virs.2022.11.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
The recently discovered SARS-CoV-2 variant Omicron (B.1.1.529) has rapidly become a global public health issue. The substantial mutations in the spike protein in this new variant have raised concerns about its ability to escape from pre-existing immunity established by natural infection or vaccination. In this review, we give a summary of current knowledge concerning the antibody evasion properties of Omicron and its subvariants (BA.2, BA.2.12.1, BA.4/5, and BA.2.75) from therapeutic monoclonal antibodies and the sera of SARS-CoV-2 vaccine recipients or convalescent patients. We also summarize whether vaccine-induced cellular immunity (memory B cell and T cell response) can recognize Omicron specifically. In brief, the Omicron variants demonstrated remarkable antibody evasion, with even more striking antibody escape seen in the Omicron BA.4 and BA.5 sub-lineages. Luckily, the third booster vaccine dose significantly increased the neutralizing antibodies titers, and the vaccine-induced cellular response remains conserved and provides second-line defense against the Omicron.
Collapse
Affiliation(s)
- Tiandan Xiang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Junzhong Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xin Zheng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
29
|
Pettini E, Medaglini D, Ciabattini A. Profiling the B cell immune response elicited by vaccination against the respiratory virus SARS-CoV-2. Front Immunol 2022; 13:1058748. [PMID: 36505416 PMCID: PMC9729280 DOI: 10.3389/fimmu.2022.1058748] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
B cells play a fundamental role in host defenses against viral infections. Profiling the B cell response elicited by SARS-CoV-2 vaccination, including the generation and persistence of antigen-specific memory B cells, is essential for improving the knowledge of vaccine immune responsiveness, beyond the antibody response. mRNA-based vaccines have shown to induce a robust class-switched memory B cell response that persists overtime and is boosted by further vaccine administration, suggesting that memory B cells are critical in driving a recall response upon re-exposure to SARS-CoV-2 antigens. Here, we focus on the role of the B cell response in the context of SARS-CoV-2 vaccination, offering an overview of the different technologies that can be used to identify spike-specific B cells, characterize their phenotype using machine learning approaches, measure their capacity to reactivate following antigen encounter, and tracking the maturation of the B cell receptor antigenic affinity.
Collapse
Affiliation(s)
| | | | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
30
|
Zhu Q, Xu Y, Wang T, Xie F. Innate and adaptive immune response in SARS-CoV-2 infection-Current perspectives. Front Immunol 2022; 13:1053437. [PMID: 36505489 PMCID: PMC9727711 DOI: 10.3389/fimmu.2022.1053437] [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: 09/25/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been a global pandemic, caused by a novel coronavirus strain with strong infectivity, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). With the in-depth research, the close relationship between COVID-19 and immune system has been dug out. During the infection, macrophages, dendritic cells, natural killer cells, CD8+ T cells, Th1, Th17, Tfh cells and effector B cells are all involved in the anti-SARS-CoV-2 responses, however, the dysfunctional immune responses will ultimately lead to the excessive inflammation, acute lung injury, even other organ failure. Thus, a detailed understanding of pertinent immune response during COVID-19 will provide insights in predicting disease outcomes and developing appropriate therapeutic approaches. In this review, we mainly clarify the role of immune cells in COVID-19 and the target-vaccine development and treatment.
Collapse
Affiliation(s)
- Qiugang Zhu
- Department of Laboratory Medicine, Shangyu People’s Hospital of Shaoxing, Shaoxing, China
| | - Yan Xu
- Department of Respiratory Medicine, Shangyu People’s Hospital of Shaoxing, Shaoxing, China
| | - Ting Wang
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Feiting Xie
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,*Correspondence: Feiting Xie,
| |
Collapse
|
31
|
Li Z, Chen X, Dan J, Hu T, Hu Y, Liu S, Chai Y, Shi Y, Wu J, Ni H, Zhu J, Wu Y, Li N, Yu Y, Wang Z, Zhao J, Zhong N, Ren X, Shen Z, Cao X. Innate immune imprints in SARS-CoV-2 Omicron variant infection convalescents. Signal Transduct Target Ther 2022; 7:377. [PMID: 36379915 PMCID: PMC9666472 DOI: 10.1038/s41392-022-01237-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
SARS-CoV-2 Omicron variant infection generally gives rise to asymptomatic to moderate COVID-19 in vaccinated people. The immune cells can be reprogrammed or "imprinted" by vaccination and infections to generate protective immunity against subsequent challenges. Considering the immune imprint in Omicron infection is unclear, here we delineate the innate immune landscape of human Omicron infection via single-cell RNA sequencing, surface proteome profiling, and plasma cytokine quantification. We found that monocyte responses predominated in immune imprints of Omicron convalescents, with IL-1β-associated and interferon (IFN)-responsive signatures with mild and moderate symptoms, respectively. Low-density neutrophils increased and exhibited IL-1β-associated and IFN-responsive signatures similarly. Mild convalescents had increased blood IL-1β, CCL4, IL-9 levels and PI3+ neutrophils, indicating a bias to IL-1β responsiveness, while moderate convalescents had increased blood CXCL10 and IFN-responsive monocytes, suggesting durative IFN responses. Therefore, IL-1β- or IFN-responsiveness of myeloid cells may indicate the disease severity of Omicron infection and mediate post-COVID conditions.
Collapse
Affiliation(s)
- Zhiqing Li
- grid.73113.370000 0004 0369 1660National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai, 200433 China
| | - Xiaosu Chen
- grid.216938.70000 0000 9878 7032Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Junyan Dan
- grid.73113.370000 0004 0369 1660National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai, 200433 China
| | - Tianju Hu
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Basic Medical Research, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005 China
| | - Ye Hu
- grid.216938.70000 0000 9878 7032Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Shuxun Liu
- grid.73113.370000 0004 0369 1660National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai, 200433 China
| | - Yangyang Chai
- grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Basic Medical Research, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005 China
| | - Yansong Shi
- grid.216938.70000 0000 9878 7032Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Jian Wu
- grid.73113.370000 0004 0369 1660National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai, 200433 China
| | - Hailai Ni
- grid.411525.60000 0004 0369 1599The Health Care Department, Shanghai Changhai Hospital, Shanghai, 200433 China
| | - Jiaqi Zhu
- grid.411525.60000 0004 0369 1599Department of Cardiology, Shanghai Changhai Hospital, Shanghai, 200433 China
| | - Yanfeng Wu
- grid.73113.370000 0004 0369 1660National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai, 200433 China
| | - Nan Li
- grid.73113.370000 0004 0369 1660National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai, 200433 China
| | - Yizhi Yu
- grid.73113.370000 0004 0369 1660National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai, 200433 China
| | | | - Jincun Zhao
- Guangzhou Laboratory, Guangzhou, 510300 China
| | | | | | - Zhongyang Shen
- grid.216938.70000 0000 9878 7032Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, 300192 China
| | - Xuetao Cao
- grid.73113.370000 0004 0369 1660National Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University, Shanghai, 200433 China ,grid.216938.70000 0000 9878 7032Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, 300071 China ,grid.506261.60000 0001 0706 7839Department of Immunology, Institute of Basic Medical Research, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005 China
| |
Collapse
|
32
|
Chen Q, Zhang J, Wang P, Zhang Z. The mechanisms of immune response and evasion by the main SARS-CoV-2 variants. iScience 2022; 25:105044. [PMID: 36068846 PMCID: PMC9436868 DOI: 10.1016/j.isci.2022.105044] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic. SARS-CoV-2 carries a unique group of mutations, and the transmission of the virus has led to the emergence of other mutants such as Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Kappa (B.1.617.1), Delta (B.1.617.2) and Omicron (B.1.1.529). The advent of a vaccine has raised hopes of ending the pandemic. However, the mutation variants of SARS-CoV-2 have raised concerns about the effectiveness of vaccines because the data showed that the vaccine was less effective against mutation variants compared to the previous variants. Mutation variants could easily mutate the N-segment structure and receptor domain of its spike glycoprotein (S) protein to escape antibody recognition. Therefore, it is vital to understand the potential immune response and evasion mechanism of SARS-CoV-2 variants. In this review, immune response and evasion mechanisms of several SARS-CoV-2 variants are described, which could provide some helpful advice for future vaccines.
Collapse
Affiliation(s)
- Qiuli Chen
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, Zhejiang 310018, China
| | - Jiawei Zhang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Peter Wang
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, Zhejiang 310018, China
| | - Zuyong Zhang
- The Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310023, China
| |
Collapse
|
33
|
Hide and seek with SARS-CoV-2: spike receptor-binding domain-specific memory B cells still recognize Omicron variant. Signal Transduct Target Ther 2022; 7:343. [PMID: 36184648 PMCID: PMC9526784 DOI: 10.1038/s41392-022-01192-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/03/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
|
34
|
Qi F, Cao Y, Zhang S, Zhang Z. Single-cell analysis of the adaptive immune response to SARS-CoV-2 infection and vaccination. Front Immunol 2022; 13:964976. [PMID: 36119105 PMCID: PMC9478577 DOI: 10.3389/fimmu.2022.964976] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/10/2022] [Indexed: 12/04/2022] Open
Abstract
Amid the ongoing Coronavirus Disease 2019 (COVID-19) pandemic, vaccination and early therapeutic interventions are the most effective means to combat and control the severity of the disease. Host immune responses to SARS-CoV-2 and its variants, particularly adaptive immune responses, should be fully understood to develop improved strategies to implement these measures. Single-cell multi-omic technologies, including flow cytometry, single-cell transcriptomics, and single-cell T-cell receptor (TCR) and B-cell receptor (BCR) profiling, offer a better solution to examine the protective or pathological immune responses and molecular mechanisms associated with SARS-CoV-2 infection, thus providing crucial support for the development of vaccines and therapeutics for COVID-19. Recent reviews have revealed the overall immune landscape of natural SARS-CoV-2 infection, and this review will focus on adaptive immune responses (including T cells and B cells) to SARS-CoV-2 revealed by single-cell multi-omics technologies. In addition, we explore how the single-cell analyses disclose the critical components of immune protection and pathogenesis during SARS-CoV-2 infection through the comparison between the adaptive immune responses induced by natural infection and by vaccination.
Collapse
Affiliation(s)
- Furong Qi
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Single-Cell Omics Reasearch and Application, Shenzhen, China
| | - Yingyin Cao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Shuye Zhang
- Clinical Center for BioTherapy and Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Single-Cell Omics Reasearch and Application, Shenzhen, China
- Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, China
| |
Collapse
|
35
|
Kaku CI, Bergeron AJ, Ahlm C, Normark J, Sakharkar M, Forsell MNE, Walker LM. Recall of preexisting cross-reactive B cell memory after Omicron BA.1 breakthrough infection. Sci Immunol 2022; 7:eabq3511. [PMID: 35549299 PMCID: PMC9097882 DOI: 10.1126/sciimmunol.abq3511] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/06/2022] [Indexed: 12/13/2022]
Abstract
Understanding immune responses after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) breakthrough infection will facilitate the development of next-generation vaccines. Here, we profiled spike (S)-specific B cell responses after Omicron/BA.1 infection in messenger RNA-vaccinated donors. The acute antibody response was characterized by high levels of somatic hypermutation and a bias toward recognition of ancestral SARS-CoV-2 strains, suggesting the early activation of vaccine-induced memory B cells. BA.1 breakthrough infection induced a shift in B cell immunodominance hierarchy from the S2 subunit, which is highly conserved across SARS-CoV-2 variants of concern (VOCs), and toward the antigenically variable receptor binding domain (RBD). A large proportion of RBD-directed neutralizing antibodies isolated from BA.1 breakthrough infection donors displayed convergent sequence features and broadly recognized SARS-CoV-2 VOCs. Together, these findings provide insights into the role of preexisting immunity in shaping the B cell response to heterologous SARS-CoV-2 variant exposure.
Collapse
Affiliation(s)
| | - Alan J. Bergeron
- Norris Cotton Cancer Center, Dartmouth Hitchcock Medical Center, Lebanon, NH 03766, USA
- Department of Microbiology and Immunology, Dartmouth College, Hanover, NH 03755, USA
| | - Clas Ahlm
- Division of Immunology, Department of Clinical Microbiology, Umea University, Umea
| | - Johan Normark
- Division of Immunology, Department of Clinical Microbiology, Umea University, Umea
| | | | | | | |
Collapse
|
36
|
Lyke KE, Atmar RL, Islas CD, Posavad CM, Szydlo D, Paul Chourdhury R, Deming ME, Eaton A, Jackson LA, Branche AR, El Sahly HM, Rostad CA, Martin JM, Johnston C, Rupp RE, Mulligan MJ, Brady RC, Frenck RW, Bäcker M, Kottkamp AC, Babu TM, Rajakumar K, Edupuganti S, Dobrzynski D, Coler RN, Archer JI, Crandon S, Zemanek JA, Brown ER, Neuzil KM, Stephens DS, Post DJ, Nayak SU, Suthar MS, Roberts PC, Beigel JH, Montefiori DC. Rapid decline in vaccine-boosted neutralizing antibodies against SARS-CoV-2 Omicron variant. Cell Rep Med 2022; 3:100679. [PMID: 35798000 PMCID: PMC9212999 DOI: 10.1016/j.xcrm.2022.100679] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/29/2022] [Accepted: 06/14/2022] [Indexed: 11/27/2022]
Abstract
The Omicron variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exhibits reduced susceptibility to vaccine-induced neutralizing antibodies, requiring a boost to generate protective immunity. We assess the magnitude and short-term durability of neutralizing antibodies after homologous and heterologous boosting with mRNA and Ad26.COV2.S vaccines. All prime-boost combinations substantially increase the neutralization titers to Omicron, although the boosted titers decline rapidly within 2 months from the peak response compared with boosted titers against the prototypic D614G variant. Boosted Omicron neutralization titers are substantially higher for homologous mRNA vaccine boosting, and for heterologous mRNA and Ad26.COV2.S vaccine boosting, compared with homologous Ad26.COV2.S boosting. Homologous mRNA vaccine boosting generates nearly equivalent neutralizing activity against Omicron sublineages BA.1, BA.2, and BA.3 but modestly reduced neutralizing activity against BA.2.12.1 and BA.4/BA.5 compared with BA.1. These results have implications for boosting requirements to protect against Omicron and future variants of SARS-CoV-2. This trial was conducted under ClincalTrials.gov: NCT04889209.
Collapse
Affiliation(s)
- Kirsten E Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Robert L Atmar
- Departments of Medicine and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, USA.
| | - Clara Dominguez Islas
- Vaccine and Infectious Disease Division, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Christine M Posavad
- Vaccine and Infectious Disease Division, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Departments of Laboratory Medicine and Pathology, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Daniel Szydlo
- Statistical Center for HIV/AIDS Research and Prevention (SCHARP), University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Rahul Paul Chourdhury
- Statistical Center for HIV/AIDS Research and Prevention (SCHARP), University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Meagan E Deming
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Amanda Eaton
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Lisa A Jackson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Angela R Branche
- Department of Medicine, Division of Infectious Diseases, University of Rochester, Rochester, NY, USA
| | - Hana M El Sahly
- Departments of Medicine and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Christina A Rostad
- Department of Pediatrics and Center for Childhood Infections and Vaccines, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Judith M Martin
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Christine Johnston
- Vaccine and Infectious Disease Division, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Departments of Laboratory Medicine and Pathology, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA
| | - Richard E Rupp
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Mark J Mulligan
- NYU Langone Vaccine Center and Division of Infectious Diseases and Immunology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Rebecca C Brady
- Cincinnati Children's Hospital Medical Center, Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Robert W Frenck
- Cincinnati Children's Hospital Medical Center, Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Martín Bäcker
- NYU Langone Hospital-Long Island Vaccine Center Research Clinic and Division of Infectious Disease, Department of Medicine, NYU Long Island School of Medicine, Mineola, NY, USA
| | - Angelica C Kottkamp
- NYU Langone Vaccine Center and Division of Infectious Diseases and Immunology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Tara M Babu
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kumaravel Rajakumar
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Srilatha Edupuganti
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA; Hope Clinic of Emory Vaccine Center, Atlanta, GA, USA
| | - David Dobrzynski
- Department of Medicine, Division of Infectious Diseases, University of Rochester, Rochester, NY, USA
| | - Rhea N Coler
- Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, WA, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Sonja Crandon
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jillian A Zemanek
- Statistical Center for HIV/AIDS Research and Prevention (SCHARP), University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Elizabeth R Brown
- Vaccine and Infectious Disease Division, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David S Stephens
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Diane J Post
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Seema U Nayak
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Yerkes National Primate Research Center, Department of Pediatrics, Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Paul C Roberts
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John H Beigel
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David C Montefiori
- Department of Surgery, Duke University Medical Center, Durham, NC, USA; Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
37
|
Wang Y, Tian Q, Ye L. The Differentiation and Maintenance of SARS-CoV-2-Specific Follicular Helper T Cells. Front Cell Infect Microbiol 2022; 12:953022. [PMID: 35909969 PMCID: PMC9329515 DOI: 10.3389/fcimb.2022.953022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/20/2022] [Indexed: 12/24/2022] Open
Abstract
Upon acute viral infection, virus-specific CD4+ T cells differentiate into either TH1 cells or follicular helper T (TFH) cells. The molecular pathways governing such bimodal cell fate commitment remain elusive. Additionally, effector virus-specific TFH cells further differentiate into corresponding memory population, which confer long-term protection against re-infection of same viruses by providing immediate help to virus-specific memory B cells. Currently, the molecular mechanisms underlying the long-term maintenance of memory TFH cells are largely unknown. In this review, we discuss current understanding of early differentiation of virus-specific effector TFH cells and long-term maintenance of virus-specific memory TFH cells in mouse models of viral infection and patients of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection.
Collapse
Affiliation(s)
- Yifei Wang
- Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Qin Tian
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- Institute of Immunology, The People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| | - Lilin Ye
- Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Institute of Immunology, The People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
- *Correspondence: Lilin Ye,
| |
Collapse
|
38
|
da Silva ES, Kohnen M, Gilson G, Staub T, Arendt V, Hilger C, Servais JY, Charpentier E, Domingues O, Snoeck CJ, Ollert M, Seguin-Devaux C, Perez-Bercoff D. Pre-Omicron Vaccine Breakthrough Infection Induces Superior Cross-Neutralization against SARS-CoV-2 Omicron BA.1 Compared to Infection Alone. Int J Mol Sci 2022; 23:7675. [PMID: 35887023 PMCID: PMC9320437 DOI: 10.3390/ijms23147675] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 02/05/2023] Open
Abstract
SARS-CoV-2 variants raise concern because of their high transmissibility and their ability to evade neutralizing antibodies elicited by prior infection or by vaccination. Here, we compared the neutralizing abilities of sera from 70 unvaccinated COVID-19 patients infected before the emergence of variants of concern (VOCs) and of 16 vaccine breakthrough infection (BTI) cases infected with Gamma or Delta against the ancestral B.1 strain, the Gamma, Delta and Omicron BA.1 VOCs using live virus. We further determined antibody levels against the Nucleocapsid (N) and full Spike proteins, the receptor-binding domain (RBD) and the N-terminal domain (NTD) of the Spike protein. Convalescent sera featured considerable variability in the neutralization of B.1 and in the cross-neutralization of different strains. Their neutralizing capacity moderately correlated with antibody levels against the Spike protein and the RBD. All but one convalescent serum failed to neutralize Omicron BA.1. Overall, convalescent sera from patients with moderate disease had higher antibody levels and displayed a higher neutralizing ability against all strains than patients with mild or severe forms of the disease. The sera from BTI cases fell into one of two categories: half the sera had a high neutralizing activity against the ancestral B.1 strain as well as against the infecting strain, while the other half had no or a very low neutralizing activity against all strains. Although antibody levels against the spike protein and the RBD were lower in BTI sera than in unvaccinated convalescent sera, most neutralizing sera also retained partial neutralizing activity against Omicron BA.1, suggestive of a better cross-neutralization and higher affinity of vaccine-elicited antibodies over virus-induced antibodies. Accordingly, the IC50: antibody level ratios were comparable for BTI and convalescent sera, but remained lower in the neutralizing convalescent sera from patients with moderate disease than in BTI sera. The neutralizing activity of BTI sera was strongly correlated with antibodies against the Spike protein and the RBD. Together, these findings highlight qualitative differences in antibody responses elicited by infection in vaccinated and unvaccinated individuals. They further indicate that breakthrough infection with a pre-Omicron variant boosts immunity and induces cross-neutralizing antibodies against different strains, including Omicron BA.1.
Collapse
Affiliation(s)
- Eveline Santos da Silva
- HIV Clinical and Translational Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (E.S.d.S.); (J.-Y.S.); (C.S.-D.)
| | - Michel Kohnen
- Centre Hospitalier de Luxembourg, 4 rue Ernest Barblé, L-1210 Luxembourg, Luxembourg; (M.K.); (G.G.); (T.S.); (V.A.)
| | - Georges Gilson
- Centre Hospitalier de Luxembourg, 4 rue Ernest Barblé, L-1210 Luxembourg, Luxembourg; (M.K.); (G.G.); (T.S.); (V.A.)
| | - Therese Staub
- Centre Hospitalier de Luxembourg, 4 rue Ernest Barblé, L-1210 Luxembourg, Luxembourg; (M.K.); (G.G.); (T.S.); (V.A.)
| | - Victor Arendt
- Centre Hospitalier de Luxembourg, 4 rue Ernest Barblé, L-1210 Luxembourg, Luxembourg; (M.K.); (G.G.); (T.S.); (V.A.)
| | - Christiane Hilger
- Molecular and Translational Allergology, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg;
| | - Jean-Yves Servais
- HIV Clinical and Translational Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (E.S.d.S.); (J.-Y.S.); (C.S.-D.)
| | - Emilie Charpentier
- Clinical and Applied Virology, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (E.C.); (C.J.S.)
| | - Olivia Domingues
- Allergy and Clinical Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (O.D.); (M.O.)
| | - Chantal J. Snoeck
- Clinical and Applied Virology, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (E.C.); (C.J.S.)
| | - Markus Ollert
- Allergy and Clinical Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (O.D.); (M.O.)
| | - Carole Seguin-Devaux
- HIV Clinical and Translational Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (E.S.d.S.); (J.-Y.S.); (C.S.-D.)
| | - Danielle Perez-Bercoff
- HIV Clinical and Translational Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; (E.S.d.S.); (J.-Y.S.); (C.S.-D.)
| |
Collapse
|
39
|
Attias P, Azzaoui I, El Karoui K, de La Selle A, Sokal A, Chappert P, Grimbert P, Fernandez I, Bouvier M, Samson C, Dahmane D, Rieu P, Nizard P, Fourati S, Sakhi H, Mahévas M. Immune Responses after a Third Dose of mRNA Vaccine Differ in Virus-Naive versus SARS-CoV-2- Recovered Dialysis Patients. Clin J Am Soc Nephrol 2022; 17:1008-1016. [PMID: 35764393 PMCID: PMC9269639 DOI: 10.2215/cjn.00830122] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/16/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND OBJECTIVES After two doses of mRNA vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), patients on dialysis show a defective humoral response, but a third dose could increase anti-SARS-CoV-2 spike IgG titers. Responses could be different in virus-naive and SARS-CoV-2-recovered patients on dialysis. However, characterization of memory B cell response after three doses is lacking. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We evaluated the dynamics of antireceptor binding domain IgG titers and antireceptor binding domain memory B cells until 6 months after two and three doses (administered within 6 months after the second dose) of mRNA vaccine in SARS-CoV-2-recovered and virus-naive dialysis populations. Results were analyzed by ordinary one-way ANOVA, the Kruskal-Wallis test, or the Wilcoxon matched-pairs test as appropriate. RESULTS In total, 108 individuals (59 patients on dialysis and 49 controls) were included. In virus-naive patients on dialysis, antireceptor binding domain IgG response was quantitatively lower after two doses compared with healthy controls, but IgG titers increased by three-fold after three doses (P=0.008). In SARS-CoV-2-recovered patients on dialysis, antireceptor binding domain IgG titers after two doses were significantly higher compared with virus-naive patients on dialysis but did not significantly increase after a third dose. Regarding memory B cell response, we detected receptor binding domain-specific memory B cells at similar proportions in virus-naive patients on dialysis and vaccinated controls after two doses. Moreover, a strong receptor binding domain-specific memory B cell expansion was observed after the third dose in virus-naive patients on dialysis (5.5-fold; P<0.001). However, in SARS-CoV-2-recovered patients on dialysis, antireceptor binding domain memory B cells remained unchanged after the third dose. CONCLUSIONS The third dose of mRNA vaccine given within 6 months after the second dose boosts serologic and memory response in virus-naive patients but not in SARS-CoV-2-recovered patients on dialysis. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER COVID-19: SARS-CoV-2 Specific Memory B and T-CD4+ Cells (MEMO-COV2), NCT04402892.
Collapse
Affiliation(s)
- Philippe Attias
- Department of Nephrology and Dialysis, Hôpital Privé Nord Parisien, Sarcelles, France
| | - Imane Azzaoui
- Department of Internal medicine, Centre Hospitalier Universitaire Henri Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est Créteil, Creteil, France
- Institut National de la Santé et de la Recherche Médical U955, Équipe 2, Transfusion and pathologies of the red blood cell, Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Creteil, France
| | - Khalil El Karoui
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Department of Nephrology, Centre de Référence Maladie Rare Syndrome Néphrotique Idiopathique, Fédération Hospitalo-Universitaire Innovative Therapy for Immune Disorders, Creteil, France
- Institut National de la Santé et de la Recherche Médical U955, Équipe 21, Pathophysiology of glomerular diseases, Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Creteil, France
| | - Andréa de La Selle
- Institut Necker Enfants Malades, Institut National de la Santé et de la Recherche Médical U1151/Centre National de Recherche Scientifique Unité Mixtes de Service 8253, Université de Paris, Paris, France
| | - Aurélien Sokal
- Department of Internal medicine, Centre Hospitalier Universitaire Henri Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est Créteil, Creteil, France
- Institut National de la Santé et de la Recherche Médical U955, Équipe 2, Transfusion and pathologies of the red blood cell, Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Creteil, France
| | - Pascal Chappert
- Institut Necker Enfants Malades, Institut National de la Santé et de la Recherche Médical U1151/Centre National de Recherche Scientifique Unité Mixtes de Service 8253, Université de Paris, Paris, France
- Institut Pasteur, Unité de Virologie Structurale, Centre National de Recherche Scientifique Unité Mixte de Recherche 3569, Paris, France
| | - Philippe Grimbert
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Department of Nephrology, Centre de Référence Maladie Rare Syndrome Néphrotique Idiopathique, Fédération Hospitalo-Universitaire Innovative Therapy for Immune Disorders, Creteil, France
- Institut National de la Santé et de la Recherche Médical U955, Équipe 21, Pathophysiology of glomerular diseases, Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Creteil, France
| | - Ignacio Fernandez
- Institut Pasteur, Unité de Virologie Structurale, Centre National de Recherche Scientifique Unité Mixte de Recherche 3569, Paris, France
| | - Magali Bouvier
- Département de Virologie, Bactériologie, Hygiène et Mycologie-Parasitologie, Centre Hospitalier Universitaire Henri Mondor, Assistance Publique-Hôpitaux de Paris, Creteil, France
- Institut National de la Santé et de la Recherche Médical U955, Équipe 18, Viruses, Hepatology, Cancer, Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Creteil, France
| | - Chloé Samson
- Institut National de la Santé et de la Recherche Médical U955, Équipe 21, Pathophysiology of glomerular diseases, Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Creteil, France
| | - Djamal Dahmane
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Department of Nephrology, Centre de Référence Maladie Rare Syndrome Néphrotique Idiopathique, Fédération Hospitalo-Universitaire Innovative Therapy for Immune Disorders, Creteil, France
| | - Philippe Rieu
- Department of Nephrology and Dialysis, Hôpital Privé Nord Parisien, Sarcelles, France
| | | | - Slim Fourati
- Département de Virologie, Bactériologie, Hygiène et Mycologie-Parasitologie, Centre Hospitalier Universitaire Henri Mondor, Assistance Publique-Hôpitaux de Paris, Creteil, France
- Institut National de la Santé et de la Recherche Médical U955, Équipe 18, Viruses, Hepatology, Cancer, Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Creteil, France
| | - Hamza Sakhi
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Department of Nephrology, Centre de Référence Maladie Rare Syndrome Néphrotique Idiopathique, Fédération Hospitalo-Universitaire Innovative Therapy for Immune Disorders, Creteil, France
- Institut National de la Santé et de la Recherche Médical U955, Équipe 21, Pathophysiology of glomerular diseases, Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Creteil, France
| | - Matthieu Mahévas
- Department of Internal medicine, Centre Hospitalier Universitaire Henri Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est Créteil, Creteil, France
- Institut National de la Santé et de la Recherche Médical U955, Équipe 2, Transfusion and pathologies of the red blood cell, Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Creteil, France
- Institut Necker Enfants Malades, Institut National de la Santé et de la Recherche Médical U1151/Centre National de Recherche Scientifique Unité Mixtes de Service 8253, Université de Paris, Paris, France
| |
Collapse
|
40
|
Liu C, Yan W, Shi J, Wang S, Peng A, Chen Y, Huang K. Biological Actions, Implications, and Cautions of Statins Therapy in COVID-19. Front Nutr 2022; 9:927092. [PMID: 35811982 PMCID: PMC9257176 DOI: 10.3389/fnut.2022.927092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/30/2022] [Indexed: 11/21/2022] Open
Abstract
The Coronavirus Disease 2019 (COVID-19) showed worse prognosis and higher mortality in individuals with obesity. Dyslipidemia is a major link between obesity and COVID-19 severity. Statins as the most common lipid regulating drugs have shown favorable effects in various pathophysiological states. Importantly, accumulating observational studies have suggested that statin use is associated with reduced risk of progressing to severe illness and in-hospital death in COVID-19 patients. Possible explanations underlie these protective impacts include their abilities of reducing cholesterol, suppressing viral entry and replication, anti-inflammation and immunomodulatory effects, as well as anti-thrombosis and anti-oxidative properties. Despite these benefits, statin therapies have side effects that should be considered, such as elevated creatinine kinase, liver enzyme and serum glucose levels, which are already elevated in severe COVID-19. Concerns are also raised whether statins interfere with the efficacy of COVID-19 vaccines. Randomized controlled trials are being conducted worldwide to confirm the values of statin use for COVID-19 treatment. Generally, the results suggest no necessity to discontinue statin use, and no evidence suggesting interference between statins and COVID-19 vaccines. However, concomitant administration of statins and COVID-19 antiviral drug Paxlovid may increase statin exposure and the risk of adverse effects, because most statins are metabolized mainly through CYP3A4 which is potently inhibited by ritonavir, a major component of Paxlovid. Therefore, more clinical/preclinical studies are still warranted to understand the benefits, harms and mechanisms of statin use in the context of COVID-19.
Collapse
Affiliation(s)
- Chengyu Liu
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanyao Yan
- Department of Pharmacy, Wuhan Fourth Hospital, Wuhan, China
| | - Jiajian Shi
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shun Wang
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anlin Peng
- Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Yuchen Chen
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Tongji-Rongcheng Center for Biomedicine, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|