1
|
Mackay M, Wagner CA, Pinckney A, Cohen JA, Wallace ZS, Khosroshahi A, Sparks JA, Lord S, Saxena A, Caricchio R, Kim AH, Kamen DL, Koumpouras F, Askanase AD, Smith K, Guthridge JM, Pardo G, Mao-Draayer Y, Macwana S, McCarthy S, Sherman MA, Hamrah SD, Veri M, Walker S, York K, Tedeschi S, Wang J, Dziubla G, Castro M, Carroll R, Narpala S, Lin BC, Serebryannyy L, McDermott A, Barry WT, Goldmuntz E, McNamara J, Payne AS, Bar-Or A, Khanna D, James JA. Prospective SARS-CoV-2 Booster Vaccination in Immunosuppressant-Treated Systemic Autoimmune Disease Patients in a Randomized Controlled Trial. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.25.25324558. [PMID: 40196267 PMCID: PMC11974989 DOI: 10.1101/2025.03.25.25324558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background Autoimmune disease patients on immunosuppressants exhibit reduced humoral responses to primary COVID-19 vaccination. Booster vaccine responses and the effects of holding immunosuppression around vaccination are less studied. We evaluated the efficacy and safety of additional vaccination in mycophenolate mofetil/mycophenolic acid (MMF/MPA)-, methotrexate (MTX)-, and B cell-depleting therapy (BCDT)-treated autoimmune disease patients, including the impact of withholding MMF/MPA and MTX. Methods In this open-label, multicenter, randomized trial, 22 MMF/MPA-, 26 MTX-, and 93 BCDT-treated autoimmune disease patients with negative or suboptimal antibody responses to initial COVID-19 vaccines (BNT162b2, mRNA-1273, or AD26.COV2.S) received a homologous booster. MMF/MPA and MTX participants were randomized (1:1) to continue or withhold treatment around vaccination. The primary outcome was the change in anti-Wuhan-Hu-1 receptor-binding domain (RBD) concentrations at 4 weeks post-additional vaccination. Secondary outcomes included adverse events, COVID-19 infections, and autoimmune disease activity through 48 weeks. Results Additional vaccination increased anti-RBD concentrations in MMF/MPA and MTX patients, irrespective of whether immunosuppression was continued or withheld. BCDT-treated patients also demonstrated increased anti-RBD concentrations, albeit lower than MMF/MPA- and MTX-treated cohorts. COVID-19 infections occurred in 30-46% of participants, were predominantly mild, and included only two non-fatal hospitalizations. Additional vaccination was well-tolerated, with low frequencies of severe disease flares and adverse events. Conclusion Additional COVID-19 vaccination is effective and safe in immunosuppressant-treated autoimmune disease patients, regardless of whether MMF/MPA or MTX is withheld. Trial Registration. ClinicalTrials.gov (NCT#05000216).
Collapse
|
2
|
Ammitzbøll C, Thomsen MK, Erikstrup C, Troldborg A. Considerations for Coronavirus Disease 2019 Vaccination Among B-Cell-Depleted Patients. Rheum Dis Clin North Am 2025; 51:45-59. [PMID: 39550106 DOI: 10.1016/j.rdc.2024.09.006] [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] [Indexed: 11/18/2024]
Abstract
This article provides a comprehensive review of the impact of B-cell-directed therapy on severe acute respiratory syndrome coronavirus 2 vaccine immunity, focusing on its implications in autoimmune inflammatory rheumatic diseases (AIIRD). Rituximab (RTX) is the primary B-cell-depleting drug that has been studied in AIIRD and is the focus of this review. We review the pivotal role of B cells in vaccine response and propose strategies to manage and predict vaccine responses in B-cell-depleted individuals. We highlight the need to strategize patients into distinct groups when predicting vaccine responses and developing guidelines to ensure optimal outcomes for RTX-treated patients.
Collapse
Affiliation(s)
- Christian Ammitzbøll
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Christian Erikstrup
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Clinical Immunology, Aarhus University Hospital, Aarhus N, Denmark
| | - Anne Troldborg
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
3
|
Gendron N, Helley D, Thaler J, Faille D, Le Beller C, Gruest M, Hadjadj J, Philippe A, Zeco F, Courbebaisse M, Darnige L, Amara W, Calmette L, Parfait B, Auditeau C, Chocron R, Khider L, Mauge L, Espitia O, Friedlander G, Ajzenberg N, Lebeaux D, Planquette B, Sanchez O, Diehl JL, Lillo-Le Louet A, Terrier B, Smadja DM. Relevance of anti-platelet factor 4/heparin antibodies and platelet activation in systemic inflammatory diseases and thrombosis disorders: insight from the COVID-19 pandemic. Res Pract Thromb Haemost 2025; 9:102701. [PMID: 40123654 PMCID: PMC11929090 DOI: 10.1016/j.rpth.2025.102701] [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: 09/19/2024] [Revised: 12/30/2024] [Accepted: 02/01/2025] [Indexed: 03/25/2025] Open
Abstract
Background The increased interest in anti-platelet factor 4 (PF4)-heparin complex (anti-PF4/H) antibodies following the COVID-19 pandemic has established them as crucial players in immunothrombosis. Objectives We aimed to investigate the involvement of anti-PF4/H antibodies during COVID-19 and after vaccination, particularly in patients with systemic inflammatory disease (SID). Methods This retrospective study analyzed the presence of anti-PF4/H antibodies and their ability to induce platelet activation in COVID-19 patients with and without suspected heparin-induced thrombocytopenia (HIT), vaccine-induced immune thrombotic thrombocytopenia (VITT) patients, and in controls and SID patients following COVID-19 vaccination. Results No significant increase in anti-PF4/H antibody levels was observed during COVID-19 regardless of disease severity. Despite a 2-fold increase in HIT suspicion observed during the pandemic, there was no corresponding increase in HIT diagnoses. Additionally, no significant increase in anti-PF4/H levels was noted after vaccination, even in SID patients. None of the positive anti-PF4/H antibodies detected in COVID-19 or vaccination cohorts induced platelet activation, measured by soluble P-selectin levels and flow cytometry-based on platelet microvesicle generation. Finally, in VITT patients, unlike in HIT patients, anti-PF4/H levels were strongly associated with platelet microvesicle assay and moderately with soluble P-selectin levels. Conclusion Our study found no significant increase in anti-PF4/H antibodies in COVID-19 or after vaccination, including in SID patients. However, in VITT patients, but not in HIT patients, these antibodies were correlated with platelet activation. This finding suggests that anti-PF4/H antibodies play a different role in the pathophysiology of VITT but that their interest is limited outside clear contexts of HIT/VITT suspicion.
Collapse
Affiliation(s)
- Nicolas Gendron
- Hematology department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
- F-CRIN INNOVTE, Saint-Étienne, France
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
| | - Dominique Helley
- Hematology department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
| | - Johannes Thaler
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Dorothée Faille
- Paris Cité University, INSERM UMR 1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France, Laboratoire d'Hématologie, AP-HP, Bichat–Claude Bernard Hospital, Paris, France
| | - Christine Le Beller
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
- Département de Pharmacovigilance, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), Paris, France
| | - Maxime Gruest
- Hematology department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
| | - Jérôme Hadjadj
- Sorbonne Université, Service de Médecine interne, Hôpital Saint-Antoine, AP-HP, Imagine Institute, Laboratory for Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, Paris, France
| | - Aurélien Philippe
- Hematology department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
| | - Faris Zeco
- Hematology department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
| | - Marie Courbebaisse
- Université Paris Cité, Physiology Department, European Georges-Pompidou Hospital, APHP, INSERM U1151, Paris, France
| | - Luc Darnige
- Hematology department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
| | - Wafa Amara
- Hematology department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
| | - Leyla Calmette
- Hematology-Immunology-Transfusion Department, Hôpitaux Universitaires Paris Ile De France Ouest, Université Versailles Saint Quentin, Boulogne, France
| | - Beatrice Parfait
- Centre de Ressources Biologiques de l'Hôpital Cochin, AP-HP.Centre-Université Paris Cité, Paris, France
| | - Claire Auditeau
- Hematology department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
| | - Richard Chocron
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, F-75015 Paris, France, and Emergency department, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), Paris, France
| | - Lina Khider
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
- Vascular Medicine Department, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), Paris, France
| | - Laetitia Mauge
- Hematology department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
| | - Olivier Espitia
- Nantes Université, CHU Nantes, Department of Internal and Vascular Medicine, l'institut du thorax, INSERM UMR1087/CNRS UMR 6291, Team III Vascular & Pulmonary diseases, Nantes, France
| | | | - Nadine Ajzenberg
- Paris Cité University, INSERM UMR 1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France, Laboratoire d'Hématologie, AP-HP, Bichat–Claude Bernard Hospital, Paris, France
| | - David Lebeaux
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Genetics of Biofilms Laboratory, Paris, France
- Service de Microbiologie, Unité Mobile d’Infectiologie, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), Hôpital Européen Georges Pompidou, Paris, France
| | - Benjamin Planquette
- F-CRIN INNOVTE, Saint-Étienne, France
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
- Respiratory Medicine Department, Assistance Publique - Hôpitaux de Paris-Centre (APHP-CUP), Paris, France
| | - Olivier Sanchez
- F-CRIN INNOVTE, Saint-Étienne, France
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
- Respiratory Medicine Department, Assistance Publique - Hôpitaux de Paris-Centre (APHP-CUP), Paris, France
| | - Jean-Luc Diehl
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
- Intensive care medicine, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), Paris, France
| | - COVID-HOP Study Group
- Hematology department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
- F-CRIN INNOVTE, Saint-Étienne, France
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Paris Cité University, INSERM UMR 1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France, Laboratoire d'Hématologie, AP-HP, Bichat–Claude Bernard Hospital, Paris, France
- Département de Pharmacovigilance, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), Paris, France
- Sorbonne Université, Service de Médecine interne, Hôpital Saint-Antoine, AP-HP, Imagine Institute, Laboratory for Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, Paris, France
- Université Paris Cité, Physiology Department, European Georges-Pompidou Hospital, APHP, INSERM U1151, Paris, France
- Hematology-Immunology-Transfusion Department, Hôpitaux Universitaires Paris Ile De France Ouest, Université Versailles Saint Quentin, Boulogne, France
- Centre de Ressources Biologiques de l'Hôpital Cochin, AP-HP.Centre-Université Paris Cité, Paris, France
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, F-75015 Paris, France, and Emergency department, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), Paris, France
- Vascular Medicine Department, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), Paris, France
- Nantes Université, CHU Nantes, Department of Internal and Vascular Medicine, l'institut du thorax, INSERM UMR1087/CNRS UMR 6291, Team III Vascular & Pulmonary diseases, Nantes, France
- Fondation Université Paris Cité, Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Genetics of Biofilms Laboratory, Paris, France
- Service de Microbiologie, Unité Mobile d’Infectiologie, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), Hôpital Européen Georges Pompidou, Paris, France
- Respiratory Medicine Department, Assistance Publique - Hôpitaux de Paris-Centre (APHP-CUP), Paris, France
- Intensive care medicine, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), Paris, France
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, F-75015 Paris, France, Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), Paris, France
| | - Agnès Lillo-Le Louet
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
- Département de Pharmacovigilance, Assistance Publique Hôpitaux de Paris.Centre-Université de Paris (APHP-CUP), Paris, France
| | - Benjamin Terrier
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, F-75015 Paris, France, Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), Paris, France
| | - David M. Smadja
- Hematology department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Paris, France
- F-CRIN INNOVTE, Saint-Étienne, France
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
| |
Collapse
|
4
|
Geldof J, Truyens M, Sabino J, Ferrante M, Lambert J, Lapeere H, Hillary T, Van Laethem A, de Vlam K, Verschueren P, Lobaton T, Padalko E, Vermeire S. Did We Overreact? Insights on COVID-19 Disease and Vaccination in a Large Cohort of Immune-Mediated Inflammatory Disease Patients during Sequential Phases of the Pandemic (The BELCOMID Study). Vaccines (Basel) 2024; 12:1157. [PMID: 39460324 PMCID: PMC11510991 DOI: 10.3390/vaccines12101157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
INTRODUCTION As the COVID-19 pandemic becomes an endemic state, still many questions remain regarding the risks and impact of SARS-CoV-2 infection and vaccination in patients with immune-mediated inflammatory diseases (IMIDs) who were excluded from the phase 3 COVID-19 vaccination trials. METHODS The BELCOMID study collected patient data and serological samples from a large, multicentric IMID patient cohort that was prospectively followed during sequential stages of the pandemic. Patients were stratified according to vaccination status into five groups across three sampling periods. Interactions between SARS-CoV-2 infection, COVID-19 vaccination status, IMID-treatment modalities and IMID course were explored. RESULTS In total, 2165 patients with IBD, a dermatological or rheumatological IMID participated. SARS-CoV-2 infection rates increased over the course of the pandemic and were highest in IMID patients that had refused every vaccine. After baseline COVID-19 vaccination, serologic spike (S)-antibody responses were attenuated by particular types of immune-modulating treatment: anti-TNF, rituximab, JAKi, systemic steroids, combined biologic/immunomodulator treatment. Nonetheless, S-antibody concentration increased progressively in patients who received a booster vaccination, reaching 100% seroconversion rate in patients who had received two booster vaccines. Previous SARS-CoV-2 infection was found as a predictor of higher S-antibody response. Patients who had refused every vaccine showed the lowest rates of S-seroconversion (53.8%). Multiple logistic regression did not identify previous SARS-CoV-2 infection as a risk factor for IMID flare-up. Furthermore, no increased risk of IMID flare-up was found with booster vaccination. CONCLUSIONS Altogether, the BELCOMID study provides evidence for the efficacy and safety of COVID-19 vaccination and confirms the importance of repeated booster vaccination in IMID patients.
Collapse
Affiliation(s)
- Jeroen Geldof
- Department of Gastroenterology and Hepatology, Ghent University Hospital, 9000 Ghent, Belgium; (M.T.); (T.L.)
- Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Marie Truyens
- Department of Gastroenterology and Hepatology, Ghent University Hospital, 9000 Ghent, Belgium; (M.T.); (T.L.)
- Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - João Sabino
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium; (J.S.); (M.F.); (S.V.)
- Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
| | - Marc Ferrante
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium; (J.S.); (M.F.); (S.V.)
- Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
| | - Jo Lambert
- Department of Dermatology, Ghent University Hospital, 9000 Ghent, Belgium; (J.L.); (H.L.)
| | - Hilde Lapeere
- Department of Dermatology, Ghent University Hospital, 9000 Ghent, Belgium; (J.L.); (H.L.)
| | - Tom Hillary
- Department of Dermatology, University Hospitals Leuven, 3000 Leuven, Belgium; (T.H.); (A.V.L.)
| | - An Van Laethem
- Department of Dermatology, University Hospitals Leuven, 3000 Leuven, Belgium; (T.H.); (A.V.L.)
| | - Kurt de Vlam
- Department of Rheumatology, University Hospitals Leuven, 3000 Leuven, Belgium; (K.d.V.); (P.V.)
| | - Patrick Verschueren
- Department of Rheumatology, University Hospitals Leuven, 3000 Leuven, Belgium; (K.d.V.); (P.V.)
| | - Triana Lobaton
- Department of Gastroenterology and Hepatology, Ghent University Hospital, 9000 Ghent, Belgium; (M.T.); (T.L.)
- Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Elizaveta Padalko
- Department of Laboratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Séverine Vermeire
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium; (J.S.); (M.F.); (S.V.)
- Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
5
|
Kobashi Y, Kawamura T, Shimazu Y, Kaneko Y, Nishikawa Y, Sugiyama A, Tani Y, Nakayama A, Yoshida M, Zho T, Yamamoto C, Saito H, Takita M, Wakui M, Kodama T, Tsubokura M. Kinetics of humoral and cellular immune responses 5 months post-COVID-19 booster dose by immune response groups at the peak immunity phase: An observational historical cohort study using the Fukushima vaccination community survey. Vaccine X 2024; 20:100553. [PMID: 39309610 PMCID: PMC11416657 DOI: 10.1016/j.jvacx.2024.100553] [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: 05/24/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024] Open
Abstract
Background Understanding the waning of immunity after booster vaccinations is important to identify which immune-low populations should be prioritized. Methods We investigated longitudinal cellular and humoral immunity after the third vaccine dose in both high- and low-cellular and humoral immunity groups at the peak immunity phase after the booster vaccination in a large community-based cohort. Blood samples were collected from 1045 participants at peak (T1: median 54 days post-third dose) and decay (T2: median 145 days post-third dose) phases to assess IgG(S), neutralizing activity, and ELISpot responses. Participants were categorized into high/low ELISpot/IgG(S) groups at T1. Cellular and humoral responses were tracked for approximately five months after the third vaccination. Results In total, 983 participants were included in the cohort. IgG(S) geometric mean fold change between timepoints revealed greater waning in the >79 years age group (T2/T1 fold change: 0.27) and higher IgG(S) fold change in the low-ELISpot group at T1 (T2/T1 fold change: 0.32-0.33) than in the other groups, although ELISpot geometric mean remained stable. Conclusions Antibody level of those who did not respond well to third dose vaccination waned rapidly than those who responded well. Evidence-based vaccine strategies are essential in preventing potential health issues caused by vaccines, including side-effects.
Collapse
Affiliation(s)
- Yurie Kobashi
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Ishikawa District, Fukushima, Japan
| | - Takeshi Kawamura
- Isotope Science Center, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Yuzo Shimazu
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Yudai Kaneko
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
- Medical & Biological Laboratories Co., Ltd, Tokyo, Japan
| | - Yoshitaka Nishikawa
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Ishikawa District, Fukushima, Japan
| | - Akira Sugiyama
- Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Yuta Tani
- Medical Governance Research Institute, Minato-ku, Tokyo, Japan
| | - Aya Nakayama
- Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Makoto Yoshida
- Medical Governance Research Institute, Minato-ku, Tokyo, Japan
| | - Tianchen Zho
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Chika Yamamoto
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Hiroaki Saito
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Morihito Takita
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Masatoshi Wakui
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuhiko Kodama
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Masaharu Tsubokura
- Department of Radiation Health Management, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
- Department of General Internal Medicine, Hirata Central Hospital, Hirata, Ishikawa District, Fukushima, Japan
- General Incorporated Association for Comprehensive Disaster Health Management Research Institute, Japan
| |
Collapse
|
6
|
Zhou H, Leng P, Wang Y, Yang K, Li C, Ojcius DM, Wang P, Jiang S. Development of T cell antigen-based human coronavirus vaccines against nAb-escaping SARS-CoV-2 variants. Sci Bull (Beijing) 2024; 69:2456-2470. [PMID: 38942698 DOI: 10.1016/j.scib.2024.02.041] [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: 10/07/2023] [Revised: 12/15/2023] [Accepted: 02/07/2024] [Indexed: 06/30/2024]
Abstract
Currently approved vaccines have been successful in preventing the severity of COVID-19 and hospitalization. These vaccines primarily induce humoral immune responses; however, highly transmissible and mutated variants, such as the Omicron variant, weaken the neutralization potential of the vaccines, thus, raising serious concerns about their efficacy. Additionally, while neutralizing antibodies (nAbs) tend to wane more rapidly than cell-mediated immunity, long-lasting T cells typically prevent severe viral illness by directly killing infected cells or aiding other immune cells. Importantly, T cells are more cross-reactive than antibodies, thus, highly mutated variants are less likely to escape lasting broadly cross-reactive T cell immunity. Therefore, T cell antigen-based human coronavirus (HCoV) vaccines with the potential to serve as a supplementary weapon to combat emerging SARS-CoV-2 variants with resistance to nAbs are urgently needed. Alternatively, T cell antigens could also be included in B cell antigen-based vaccines to strengthen vaccine efficacy. This review summarizes recent advancements in research and development of vaccines containing T cell antigens or both T and B cell antigens derived from proteins of SARS-CoV-2 variants and/or other HCoVs based on different vaccine platforms.
Collapse
Affiliation(s)
- Hao Zhou
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400016, China.
| | - Ping Leng
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400016, China
| | - Yang Wang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Kaiwen Yang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chen Li
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - David M Ojcius
- Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, CA 94115, USA
| | - Pengfei Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai Institute of Infectious Disease and Biosecurity, School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministry of Education/Ministry of Health/Chinese Academy of Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
7
|
Hamad Saied M, van Straalen JW, de Roock S, Verduyn Lunel FM, de Wit J, de Rond LGH, Van Nieuwenhove E, Vastert BJ, van Montfrans JM, van Royen-Kerkhof A, de Joode-Smink GCJ, Swart JF, Wulffraat NM, Jansen MHA. Humoral and cellular immunogenicity, effectiveness and safety of COVID-19 mRNA vaccination in patients with pediatric rheumatic diseases: A prospective cohort study. Vaccine 2024; 42:1145-1153. [PMID: 38262809 DOI: 10.1016/j.vaccine.2024.01.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 01/02/2024] [Accepted: 01/16/2024] [Indexed: 01/25/2024]
Abstract
OBJECTIVES To evaluate immunogenicity, effectiveness and safety of COVID-19 vaccination in patients with pediatric autoimmune inflammatory rheumatic disease (pedAIIRD). METHODS A prospective cohort study was performed at the pediatric rheumatology department of the Wilhelmina Children's Hospital in Utrecht, the Netherlands. Vaccination dates, COVID-19 cases and vaccine-related adverse events (AEs) were registered for all pedAIIRD patients during regular clinic visits from March 2021 - August 2022. SARS-CoV-2 IgG antibody levels and T-cell responses were measured from serum samples after vaccination, and clinical and drug therapy data were collected from electronic medical records. Rate of COVID-19 disease was compared between vaccinated and unvaccinated patients in a time-varying Cox regression analysis. RESULTS A total of 157 patients were included in this study and 88 % had juvenile idiopathic arthritis (JIA). One hundred thirty-seven patients were fully vaccinated, of which 47 % used biological agents at the time of vaccination, and 20 patients were unvaccinated. Geometric mean concentrations (GMCs) of post-vaccine antibody levels against SARS-CoV-2 were above the threshold for positivity in patients who did and did not use biological agents at the time of vaccination, although biological users demonstrated significantly lower antibody levels (adjusted GMC ratio: 0.38, 95 % CI: 0.21 - 0.70). T-cell responses were adequate in all but two patients (9 %). The adjusted rate of reported COVID-19 was significantly lower for fully vaccinated patients compared to non-vaccinated patients (HR: 0.53, 95 % CI: 0.29 - 0.97). JIA disease activity scores were not significantly different after vaccination, and no serious AEs were reported. CONCLUSIONS COVID-19 mRNA vaccines were immunogenic (both cellular and humoral), effective and safe in a large cohort of pedAIIRD patients despite their use of immunosuppressive medication.
Collapse
Affiliation(s)
- Mohamad Hamad Saied
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Pediatrics, Carmel Medical Center, Technion Faculty of Medicine, Haifa, Israel.
| | - Joeri W van Straalen
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sytze de Roock
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Frans M Verduyn Lunel
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jelle de Wit
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Lia G H de Rond
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Erika Van Nieuwenhove
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Bas J Vastert
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands; Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joris M van Montfrans
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Annet van Royen-Kerkhof
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Gerrie C J de Joode-Smink
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joost F Swart
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Nico M Wulffraat
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marc H A Jansen
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
8
|
Delestre F, Charles P, Karras A, Pagnoux C, Néel A, Cohen P, Aumaître O, Faguer S, Gobert P, Maurier F, Samson M, Godmer P, Bonnotte B, Cottin V, Hanrotel-Saliou C, Le Gallou T, Carron PL, Desmurs-Clavel H, Direz G, Jourde-Chiche N, Lifermann F, Martin-Silva N, Pugnet G, Quéméneur T, Matignon M, Benhamou Y, Daugas E, Lazaro E, Limal N, Ducret M, Huart A, Viallard JF, Hachulla E, Perrodeau E, Puechal X, Guillevin L, Porcher R, Terrier B. Rituximab as maintenance therapy for ANCA-associated vasculitides: pooled analysis and long-term outcome of 277 patients included in the MAINRITSAN trials. Ann Rheum Dis 2024; 83:233-241. [PMID: 37918894 DOI: 10.1136/ard-2023-224623] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/14/2023] [Indexed: 11/04/2023]
Abstract
OBJECTIVE To compare the long-term efficacy and safety of azathioprine (AZA), 18-month fixed-schedule rituximab (RTX), 18-month tailored RTX and 36-month RTX in preventing relapses in patients with antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis who achieved a complete remission after induction therapy. Patients treated with 36-month RTX received either a fixed or a tailored regimen for the first 18 months and a fixed regimen for the last 18 months (36-month fixed/fixed RTX and 36-month tailored/fixed RTX, respectively). METHODS The Maintenance of Remission using Rituximab in Systemic ANCA-associated Vasculitis (MAINRITSAN) trials sequentially compared: 18-month fixed-schedule RTX versus AZA (MAINRITSAN); 18-month fixed-schedule RTX versus 18-month tailored-RTX (MAINRITSAN2); and extended therapy to 36 months with four additional RTX infusions after MAINRITSAN2 versus placebo (MAINRITSAN3). Patients were then followed prospectively through month 84 and their data were pooled to analyse relapses and adverse events. The primary endpoint was relapse-free survival at month 84. RESULTS 277 patients were enrolled and divided in 5 groups: AZA (n=58), 18-month fixed-schedule RTX (n=97), 18-month tailored-RTX (n=40), 36-month tailored/fixed RTX (n=42), 36-month fixed/fixed RTX (n=41). After adjustment for prognostic factors, 18-month fixed-schedule RTX was superior to AZA in preventing major relapses at month 84 (HR 0.38, 95% CI 0.20 to 0.71). The 18-month tailored-RTX regimen was associated with an increased risk of major relapse compared with fixed-schedule regimen (HR 2.92, 95% CI 1.43 to 5.96). The risk of major relapse was similar between 36-month fixed/fixed and 18-month fixed-RTX (HR 0.69, 95% CI 0.38 to 1.25). CONCLUSIONS According to these results, it appears that the 84-month remission rate is higher with an 18-month fixed RTX regimen compared with AZA and 18-month tailored RTX. Also, extending RTX to 36 months does not appear to reduce the long-term relapse rate compared with the 18-month fixed RTX regimen. However, as this study was underpowered to make this comparison, further prospective studies are needed to determine the potential long-term benefits of extending treatment in these patients.
Collapse
Affiliation(s)
- Florence Delestre
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP.Centre, Hospital Cochin, Paris, France
- Université Paris Cité, Paris, France
| | - Pierre Charles
- Université Paris Cité, Paris, France
- Department of Internal Medicine, Institut Mutualiste Montsouris, Paris, France
| | - Alexandre Karras
- Université Paris Cité, Paris, France
- Department of Nephrology, Hôpital Européen Georges Pompidou, APHP, Paris, France
| | - Christian Pagnoux
- University of Toronto, Toronto, Ontario, Canada
- Vasculitis clinic, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Antoine Néel
- Department of Internal Medicine, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Pascal Cohen
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP.Centre, Hospital Cochin, Paris, France
| | - Olivier Aumaître
- Department of Internal Medicine, Hôpital Gabriel Montpied, Clermont-Ferrand, France
| | - Stanislas Faguer
- Département de Néphrologie et Transplantation d'Organes, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Pierre Gobert
- Département de médecine, Hopital Général Henri-Duffaut, Avignon, France
| | - François Maurier
- Department of Internal Medicine, Hôpitaux Privés de Metz, Metz, France
| | - Maxime Samson
- Département de Médecine Interne et Immunologie Clinique, Centre Hospitalier Universitaire de Dijon, Dijon, France
| | - Pascal Godmer
- Département de Hématologie Immunologie, Centre Hospitalier Bretagne Atlantique de Vannes, Vannes, France
| | - Bernard Bonnotte
- Département de Médecine Interne et Immunologie Clinique, Centre Hospitalier Universitaire de Dijon, Dijon, France
| | - Vincent Cottin
- Department of Respiratory Medicine, National Coordinating Reference Center for Rare Pulmonary Diseases, Louis Pradel Hospital, Lyon, France
| | - Catherine Hanrotel-Saliou
- Department of Nephrology, Centre Hospitalier Universitaire de Brest, Hôpital la Cavale Blanche, Brest, France
| | - Thomas Le Gallou
- Department of Internal Medicine, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Pierre-Louis Carron
- Département de néphrologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | | | - Guillaume Direz
- Rheumatology Department, Le Mans General Hospital, Le Mans, France
| | - Noémie Jourde-Chiche
- Centre de Néphrologie et Transplantation Rénale, Hôpital de La Conception, Aix-Marseille Université, Marseille, France
| | | | - Nicolas Martin-Silva
- Department of Internal Medicine, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Grégory Pugnet
- Department of Internal Medicine, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Thomas Quéméneur
- Département de Néphrologie et de Médecine Interne, Centre Hospitalier de Valenciennes, Valenciennes, France
| | - Marie Matignon
- Nephrology and Renal Transplantation Department, Hopitaux Universitaires Henri Mondor, Créteil, France
| | - Ygal Benhamou
- Department of Internal Medicine, Centre Hospitalier Universitaire Charles Nicolle, Rouen, France
| | - Eric Daugas
- Department of Nephrology, Hopital Bichat - Claude-Bernard, Paris, France
| | - Estibaliz Lazaro
- Department of Internal Medicine, Bordeaux University Hospital, Pessac, France
| | - Nicolas Limal
- Department of Internal Medicine, Hôpitaux Universitaires Henri Mondor, Créteil, France
| | - Maïzé Ducret
- Department of Nephrology, Annecy Hospital, Annecy, France
| | - Antoine Huart
- Department of Nephrology, Hospital Rangueil, Toulouse, France
| | | | - Eric Hachulla
- Département de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Systémiques et Auto-Immunes Rares du Nord-Ouest (CERAINO), Centre Hospitalier Universitaire de Lille, Lille, France
| | - Elodie Perrodeau
- Center of Research in Epidemiology and Statistics Sorbonne Paris Cité, Paris, France
| | - Xavier Puechal
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP.Centre, Hospital Cochin, Paris, France
- Université Paris Cité, Paris, France
| | - Loïc Guillevin
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP.Centre, Hospital Cochin, Paris, France
- Université Paris Cité, Paris, France
| | - Raphaël Porcher
- Université Paris Cité, Paris, France
- Center of Research in Epidemiology and Statistics Sorbonne Paris Cité, Paris, France
| | - Benjamin Terrier
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP.Centre, Hospital Cochin, Paris, France
- Université Paris Cité, Paris, France
| |
Collapse
|
9
|
Lingas G, Planas D, Péré H, Porrot F, Guivel-Benhassine F, Staropoli I, Duffy D, Chapuis N, Gobeaux C, Veyer D, Delaugerre C, Le Goff J, Getten P, Hadjadj J, Bellino A, Parfait B, Treluyer JM, Schwartz O, Guedj J, Kernéis S, Terrier B. Neutralizing Antibody Levels as a Correlate of Protection Against SARS-CoV-2 Infection: A Modeling Analysis. Clin Pharmacol Ther 2024; 115:86-94. [PMID: 37795693 DOI: 10.1002/cpt.3069] [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: 06/13/2023] [Accepted: 09/22/2023] [Indexed: 10/06/2023]
Abstract
Although anti-severe acute respiratory syndrome-coronavirus 2 antibody kinetics have been described in large populations of vaccinated individuals, we still poorly understand how they evolve during a natural infection and how this impacts viral clearance. For that purpose, we analyzed the kinetics of both viral load and neutralizing antibody levels in a prospective cohort of individuals during acute infection with alpha variant. Using a mathematical model, we show that the progressive increase in neutralizing antibodies leads to a shortening of the half-life of both infected cells and infectious viral particles. We estimated that the neutralizing activity reached 90% of its maximal level within 11 days after symptom onset and could reduce the half-life of both infected cells and circulating virus by a 6-fold factor, thus playing a key role to achieve rapid viral clearance. Using this model, we conducted a simulation study to predict in a more general context the protection conferred by pre-existing neutralization titers, due to either vaccination or prior infection. We predicted that a neutralizing activity, as measured by 50% effective dose > 103 , could reduce by 46% the risk of having viral load detectable by standard polymerase chain reaction assays and by 98% the risk of having viral load above the threshold of infectiousness. Our model shows that neutralizing activity could be used to define correlates of protection against infection and transmission.
Collapse
Affiliation(s)
| | - Delphine Planas
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
- Vaccine Research Institute, Créteil, France
| | - Hélène Péré
- Virology Unit, Microbiology Department, APHP, Hôpital Européen Georges-Pompidou, Paris, France
- Université Paris Cité, INSERM UMRS1138 Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Françoise Porrot
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
| | | | - Isabelle Staropoli
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Nicolas Chapuis
- Assistance Publique-Hôpitaux de Paris, Centre-Université Paris Cité, Service d'hématologie biologique, Hôpital Cochin, Paris, France
| | - Camille Gobeaux
- Department of Automated Biology, CHU de Cochin, AP-HP, Paris, France
| | - David Veyer
- Virology Unit, Microbiology Department, APHP, Hôpital Européen Georges-Pompidou, Paris, France
- Université Paris Cité, INSERM UMRS1138 Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Constance Delaugerre
- Virology Department, AP-HP, Hôpital Saint-Louis, Paris, France
- Université Paris Cité, Inserm U944, Biology of Emerging Viruses, Paris, France
| | - Jérôme Le Goff
- Virology Department, AP-HP, Hôpital Saint-Louis, Paris, France
- Université Paris Cité, Inserm U976, INSIGHT Team, Paris, France
| | | | - Jérôme Hadjadj
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP.CUP, Hôpital Cochin, Paris, France
| | - Adèle Bellino
- URC-CIC Paris Centre Necker/Cochin, AP-HP, Hôpital Cochin, Paris, France
| | - Béatrice Parfait
- Fédération des Centres de Ressources Biologiques - Plateformes de Ressources Biologiques AP-HP.Centre-Université Paris Cité, Centre de Ressources Biologiques Cochin, Hôpital Cochin, Paris, France
| | - Jean-Marc Treluyer
- Unité de Recherche clinique, Hôpital Cochin, AP-HP.Centre - Université de Paris, Paris, France
| | - Olivier Schwartz
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
- Vaccine Research Institute, Créteil, France
| | | | - Solen Kernéis
- Université Paris Cité, IAME, INSERM, Paris, France
- Equipe de Prévention du Risque Infectieux (EPRI), AP-HP, Hôpital Bichat, Paris, France
| | - Benjamin Terrier
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP.CUP, Hôpital Cochin, Paris, France
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
10
|
Shirata M, Ito I, Tanaka M, Murata K, Murakami K, Ikeda H, Oi I, Hamao N, Nishioka K, Hayashi Y, Nagao M, Hashimoto M, Ito H, Ueno H, Morinobu A, Hirai T. Impact of methotrexate on humoral and cellular immune responses to SARS-CoV-2 mRNA vaccine in patients with rheumatoid arthritis. Clin Exp Med 2023; 23:4707-4720. [PMID: 37582911 DOI: 10.1007/s10238-023-01163-5] [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: 01/24/2023] [Accepted: 07/31/2023] [Indexed: 08/17/2023]
Abstract
The aim of this study was to longitudinally evaluate the undetermined impact of methotrexate (MTX) on the cumulative immunogenicity elicited by three doses of SARS-CoV-2 mRNA vaccination in patients with rheumatoid arthritis (RA). We prospectively evaluated vaccine-induced immune responses following the first dose, 1 and 6 months after the second dose, and 1 month after the third dose of BNT162b2 or mRNA-1273 in 144 SARS-CoV-2 naïve participants (70 patients with RA, 29 disease controls without immunosuppressive conditions, and 45 healthy controls). Humoral immune responses were assessed by quantifying anti-spike IgG antibody titers and the capacity of circulating antibodies to neutralize the ancestral SARS-CoV-2 strain and the Alpha, Delta, and Omicron variants. Vaccine-induced T-cell responses were measured using an interferon-gamma release assay. At 1 and 6 months after the second dose, anti-spike titers were highest in healthy controls, followed by disease controls and patients with RA. Multivariate analyses revealed that MTX treatment was significantly associated with a decrease in anti-spike titers, neutralizing activity, and SARS-CoV-2-specific interferon-gamma levels. Furthermore, MTX dose per body weight was negatively correlated with these two indices of humoral immune response. The third vaccine dose boosted anti-spike titers, especially in patients receiving MTX, while sera from these patients neutralized the Omicron variant far less robustly than those from healthy controls. In conclusion, MTX attenuated immunogenicity following two doses of SARS-CoV-2 mRNA vaccine in patients with RA, particularly resulting in dose-dependent suppression of the humoral immune response. Furthermore, MTX deteriorated the neutralizing activity against the Omicron variant, even after the third immunization.
Collapse
Affiliation(s)
- Masahiro Shirata
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan
| | - Isao Ito
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan.
- Department of Internal Medicine, Sugita Genpaku Memorial Obama Municipal Hospital, Fukui, Japan.
| | - Masao Tanaka
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichi Murata
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kosaku Murakami
- Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Ikeda
- Department of Internal Medicine, Sugita Genpaku Memorial Obama Municipal Hospital, Fukui, Japan
| | - Issei Oi
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan
- Department of Internal Medicine, Sugita Genpaku Memorial Obama Municipal Hospital, Fukui, Japan
| | - Nobuyoshi Hamao
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan
- Department of Internal Medicine, Sugita Genpaku Memorial Obama Municipal Hospital, Fukui, Japan
| | - Kensuke Nishioka
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan
- Department of Internal Medicine, Sugita Genpaku Memorial Obama Municipal Hospital, Fukui, Japan
| | - Yasuyuki Hayashi
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan
| | - Miki Nagao
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Motomu Hashimoto
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiromu Ito
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideki Ueno
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan
| |
Collapse
|
11
|
Pappa M, Panagiotopoulos A, Thomas K, Fanouriakis A. Systemic Lupus Erythematosus and COVID-19. Curr Rheumatol Rep 2023; 25:192-203. [PMID: 37477841 PMCID: PMC10504107 DOI: 10.1007/s11926-023-01110-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2023] [Indexed: 07/22/2023]
Abstract
PURPOSE OF REVIEW To describe the current state of knowledge regarding COVID-19 in patients with systemic lupus erythematosus (SLE). We focus on (i) SARS-CoV-2 vaccination uptake, immunogenicity and safety, and (ii) outcomes of COVID-19 in patients with SLE and pertinent risk factors for adverse sequelae. RECENT FINDINGS Notwithstanding the potential concern of patients about possible post-vaccination side-effects, the safety of anti-SARS-CoV-2 vaccines in patients with SLE has been undisputedly confirmed in numerous studies. Humoral immunogenicity is generally attained in SLE, although affected by the use of background immunosuppressive drugs, especially rituximab. The latter has also clearly been implicated with adverse COVID-19 outcomes in SLE, including need for hospitalization, mechanical ventilation and death. Although the wide adoption of vaccination has significantly improved COVID-19 outcomes, patients with SLE continue to pose challenges during the pandemic, mainly owing to administered immunosuppressive medications.
Collapse
Affiliation(s)
- Maria Pappa
- 1st Department of Propaedeutic Internal Medicine, Medical School National and Kapodistrian University of Athens, Athens, Greece
| | - Alexandros Panagiotopoulos
- 1st Department of Propaedeutic Internal Medicine, Medical School National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Thomas
- 4th Department of Internal Medicine, "Attikon" University Hospital, Medical School National and Kapodistrian University of Athens, Athens, Greece
| | - Antonis Fanouriakis
- Rheumatology and Clinical Immunology, "Attikon" University Hospital of Athens, Medical School National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
12
|
Oliosi E, Flahault A, Charre C, Veyer D, Combier A, Lafont E, Karras A, Mouthon L, Avouac J, Terrier B, Hadjadj J. Impact of rituximab on humoral response to SARS-CoV-2 vaccination in previously vaccinated patients with autoimmune diseases. Clin Rheumatol 2023; 42:2485-2490. [PMID: 37243801 PMCID: PMC10224652 DOI: 10.1007/s10067-023-06638-0] [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: 02/15/2023] [Revised: 04/16/2023] [Accepted: 05/12/2023] [Indexed: 05/29/2023]
Abstract
SARS-CoV-2 infection is more severe in patients undergoing rituximab (RTX) treatment. Humoral response to vaccination is severely impaired in patients already treated with RTX, but data on antibody persistence in patients initiating RTX are lacking. We evaluated the impact of RTX initiation on humoral response to SARS-CoV-2 vaccination in previously vaccinated patients with immune-mediated inflammatory diseases. We performed a retrospective, multicenter study evaluating the evolution of anti-spike antibodies and breakthrough infections after initiation of RTX in previously vaccinated patients with protective levels of anti-SARS-CoV-2 antibodies. Threshold for anti-S antibodies positivity and protection were 30 and 264 BAU/mL, respectively. We included 31 previously vaccinated patients initiating RTX (21 female, median age 57 years). At first RTX infusion, 12 (39%) patients had received 2 doses of vaccine, 15 (48%) had received 3 doses, and 4 (13%) had received 4 doses. The most frequent underlying diseases were ANCA-associated vasculitis (29%) and rheumatoid arthritis (23%). Median anti-S antibody titers at RTX initiation, 3 months, and 6 months were 1620 (589-2080), 1055 (467-2080), and 407 (186-659) BAU/mL, respectively. Overall, antibody titers waned by almost two-fold at 3 months and four-fold at 6 months. Median antibody titers were significantly higher in patients who received ≥3 doses compared to those who received only 2 doses. Three patients developed SARS-CoV-2 infection without any severe symptom. Anti-SARS-CoV-2 antibody titers in previously vaccinated patients decline after RTX initiation similarly to general population. Specific monitoring is useful to anticipate prophylactic strategies. Key Points • Anti-SARS-CoV-2 antibody titers in previously vaccinated patients decline after rituximab initiation similarly to the general population. • The number of dose of vaccine before rituximab initiation is associated with higher antibody titers at month 3. • Monitoring antibody levels is mandatory to initiate prophylactic strategies in this population.
Collapse
Affiliation(s)
- E Oliosi
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, Hôpital Cochin, Université de Paris Cité, 75014, Paris, France.
- Service de Maladies infectieuses et tropicales, Hôpital Bicêtre, APHP, 78 rue du Général Leclerc, 94270, Le Kremlin-Bicêtre, France.
| | - A Flahault
- Department of Nephrology, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - C Charre
- Department of Virology, AP-HP, Hôpital Cochin, 75014, Paris, France
| | - D Veyer
- Department of Virology, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - A Combier
- Department of Rheumatology, AP-HP, Hôpital Cochin, 75014, Paris, France
| | - E Lafont
- Department of Internal Medicine, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - A Karras
- Department of Nephrology, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - L Mouthon
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, Hôpital Cochin, Université de Paris Cité, 75014, Paris, France
| | - J Avouac
- Department of Rheumatology, AP-HP, Hôpital Cochin, 75014, Paris, France
| | - B Terrier
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, Hôpital Cochin, Université de Paris Cité, 75014, Paris, France
| | - J Hadjadj
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, Hôpital Cochin, Université de Paris Cité, 75014, Paris, France
| |
Collapse
|
13
|
Speer C, Töllner M, Benning L, Bartenschlager M, Kim H, Nusshag C, Kälble F, Reineke M, Reichel P, Schnitzler P, Zeier M, Morath C, Schmitt W, Bergner R, Bartenschlager R, Lorenz HM, Schaier M. BA.1/BA.5 Immunogenicity, Reactogenicity, and Disease Activity after COVID-19 Vaccination in Patients with ANCA-Associated Vasculitis: A Prospective Observational Cohort Study. Viruses 2023; 15:1778. [PMID: 37632120 PMCID: PMC10458303 DOI: 10.3390/v15081778] [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: 07/07/2023] [Revised: 08/13/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023] Open
Abstract
Emerging omicron subtypes with immune escape lead to inadequate vaccine response with breakthrough infections in immunocompromised individuals such as Anti-neutrophil Cytoplasmic Antibody (ANCA)-associated vasculitis (AAV) patients. As AAV is considered an orphan disease, there are still limited data on SARS-CoV-2 vaccination and prospective studies that have focused exclusively on AAV patients are lacking. In addition, there are safety concerns regarding the use of highly immunogenic mRNA vaccines in autoimmune diseases, and further studies investigating reactogenicity are urgently needed. In this prospective observational cohort study, we performed a detailed characterization of neutralizing antibody responses against omicron subtypes and provided a longitudinal assessment of vaccine reactogenicity and AAV disease activity. Different vaccine doses were generally well tolerated and no AAV relapses occurred during follow-up. AAV patients had significantly lower anti-S1 IgG and surrogate-neutralizing antibodies after first, second, and third vaccine doses as compared to healthy controls, respectively. Live-virus neutralization assays against omicron subtypes BA.1 and BA.5 revealed that previous SARS-CoV-2 vaccines result in an inadequate neutralizing immune response in immunocompromised AAV patients. These data demonstrate that new vaccination strategies including adapted mRNA vaccines against epitopes of emerging variants are needed to help protect highly vulnerable individuals such as AAV patients.
Collapse
Affiliation(s)
- Claudius Speer
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (L.B.); (C.N.); (F.K.); (M.R.); (P.R.); (M.Z.); (C.M.); (M.S.)
- Molecular Medicine Partnership Unit Heidelberg, EMBL, 69120 Heidelberg, Germany
| | - Maximilian Töllner
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (L.B.); (C.N.); (F.K.); (M.R.); (P.R.); (M.Z.); (C.M.); (M.S.)
| | - Louise Benning
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (L.B.); (C.N.); (F.K.); (M.R.); (P.R.); (M.Z.); (C.M.); (M.S.)
| | - Marie Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, Medical Faculty Heidelberg, Heidelberg University, 68167 Heidelberg, Germany; (M.B.); (H.K.); (R.B.)
| | - Heeyoung Kim
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, Medical Faculty Heidelberg, Heidelberg University, 68167 Heidelberg, Germany; (M.B.); (H.K.); (R.B.)
| | - Christian Nusshag
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (L.B.); (C.N.); (F.K.); (M.R.); (P.R.); (M.Z.); (C.M.); (M.S.)
| | - Florian Kälble
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (L.B.); (C.N.); (F.K.); (M.R.); (P.R.); (M.Z.); (C.M.); (M.S.)
| | - Marvin Reineke
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (L.B.); (C.N.); (F.K.); (M.R.); (P.R.); (M.Z.); (C.M.); (M.S.)
| | - Paula Reichel
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (L.B.); (C.N.); (F.K.); (M.R.); (P.R.); (M.Z.); (C.M.); (M.S.)
| | - Paul Schnitzler
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Martin Zeier
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (L.B.); (C.N.); (F.K.); (M.R.); (P.R.); (M.Z.); (C.M.); (M.S.)
| | - Christian Morath
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (L.B.); (C.N.); (F.K.); (M.R.); (P.R.); (M.Z.); (C.M.); (M.S.)
| | | | - Raoul Bergner
- Department of Internal Medicine A, Clinical Center Ludwigshafen, 67071 Ludwigshafen, Germany;
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, Medical Faculty Heidelberg, Heidelberg University, 68167 Heidelberg, Germany; (M.B.); (H.K.); (R.B.)
- German Center for Infection Research (DZIF), Heidelberg Partner Site, 69120 Heidelberg, Germany
- Division Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Department of Medicine V, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Matthias Schaier
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (L.B.); (C.N.); (F.K.); (M.R.); (P.R.); (M.Z.); (C.M.); (M.S.)
| |
Collapse
|
14
|
Bruel T, Vrignaud LL, Porrot F, Staropoli I, Planas D, Guivel-Benhassine F, Puech J, Prot M, Munier S, Henry-Bolland W, Soulié C, Zafilaza K, Lusivika-Nzinga C, Meledge ML, Dorival C, Molino D, Péré H, Yordanov Y, Simon-Lorière E, Veyer D, Carrat F, Schwartz O, Marcelin AG, Martin-Blondel G. Antiviral activities of sotrovimab against BQ.1.1 and XBB.1.5 in sera of treated patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.25.23290512. [PMID: 37398037 PMCID: PMC10312842 DOI: 10.1101/2023.05.25.23290512] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Background Monoclonal antibodies (mAbs) targeting the spike of SARS-CoV-2 prevent severe COVID-19. Omicron subvariants BQ.1.1 and XBB.1.5 evade neutralization of therapeutic mAbs, leading to recommendations against their use. Yet, the antiviral activities of mAbs in treated patients remain ill-defined. Methods We investigated neutralization and antibody-dependent cellular cytotoxicity (ADCC) of D614G, BQ.1.1 and XBB.1.5 in 320 sera from 80 immunocompromised patients with mild-to-moderate COVID-19 prospectively treated with mAbs (sotrovimab, n=29; imdevimab/casirivimab, n=34; cilgavimab/tixagevimab, n=4) or anti-protease (nirmatrelvir/ritonavir, n=13). We measured live-virus neutralization titers and quantified ADCC with a reporter assay. Findings Only Sotrovimab elicits serum neutralization and ADCC against BQ.1.1 and XBB.1.5. As compared to D614G, sotrovimab neutralization titers of BQ.1.1 and XBB.1.5 are reduced (71- and 58-fold, respectively), but ADCC levels are only slightly decreased (1.4- and 1-fold, for BQ.1.1 and XBB.1.5, respectively). Interpretation Our results show that sotrovimab is active against BQ.1.1 and XBB.1.5 in treated individuals, suggesting that it may be a valuable therapeutic option.
Collapse
Affiliation(s)
- Timothée Bruel
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
- Vaccine Research Institute, Créteil, France
| | - Lou-Léna Vrignaud
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
- Sorbonne Université, Paris, France
| | - Françoise Porrot
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
| | - Isabelle Staropoli
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
| | - Delphine Planas
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
- Vaccine Research Institute, Créteil, France
| | | | - Julien Puech
- Laboratoire de Virologie, Service de Microbiologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Matthieu Prot
- G5 Evolutionary Genomics of RNA Viruses, Institut Pasteur, Université Paris Cité, Paris, France
| | - Sandie Munier
- G5 Evolutionary Genomics of RNA Viruses, Institut Pasteur, Université Paris Cité, Paris, France
| | - William Henry-Bolland
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
- École Doctorale BioSPC 562, Université de Paris, Paris, France
| | - Cathia Soulié
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et Santé Publique, 75012 Paris, France
- Virology Department, Pitié-Salpêtrière Hospital, Sorbonne University, 75013 Paris, France
| | - Karen Zafilaza
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et Santé Publique, 75012 Paris, France
- Virology Department, Pitié-Salpêtrière Hospital, Sorbonne University, 75013 Paris, France
| | - Clovis Lusivika-Nzinga
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et Santé Publique, 75012 Paris, France
| | - Marie-Laure Meledge
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et Santé Publique, 75012 Paris, France
| | - Céline Dorival
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et Santé Publique, 75012 Paris, France
| | - Diana Molino
- INSERM-ANRS Maladies Infectieuses Emergentes, 2 Oradour-Sur-Glane, 75015, Paris, France
| | - Hélène Péré
- Laboratoire de Virologie, Service de Microbiologie, Hôpital Européen Georges Pompidou, Paris, France
- Functional Genomics of Solid Tumors (FunGeST), Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Paris, France
| | - Youri Yordanov
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et Santé Publique, 75012 Paris, France
- Hôpital Saint-Antoine, Service d'Accueil des Urgences, Assistance Publique - Hôpitaux de Paris, AP-HP, Sorbonne Université, Paris, France
| | - Etienne Simon-Lorière
- G5 Evolutionary Genomics of RNA Viruses, Institut Pasteur, Université Paris Cité, Paris, France
- Institut Pasteur, Université Paris Cité, National Reference Center for viruses of respiratory infections, Paris, France
| | - David Veyer
- Laboratoire de Virologie, Service de Microbiologie, Hôpital Européen Georges Pompidou, Paris, France
- Functional Genomics of Solid Tumors (FunGeST), Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Paris, France
| | - Fabrice Carrat
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et Santé Publique, 75012 Paris, France
- Hôpital Saint-Antoine, santé publique, APHP Paris, France
| | - Olivier Schwartz
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
- Vaccine Research Institute, Créteil, France
| | - Anne-Geneviève Marcelin
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et Santé Publique, 75012 Paris, France
- Virology Department, Pitié-Salpêtrière Hospital, Sorbonne University, 75013 Paris, France
| | - Guillaume Martin-Blondel
- Service des Maladies Infectieuses et Tropicales, CHU de Toulouse, France; Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity) INSERM, Université Toulouse III., Toulouse, France
| |
Collapse
|
15
|
Kasperkiewicz M, Woodley DT. Rituximab in immunobullous disorders during the COVID-19 pandemic: A systematic review of observational studies. J Eur Acad Dermatol Venereol 2023; 37:e17-e19. [PMID: 35989595 DOI: 10.1111/jdv.18538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/18/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Michael Kasperkiewicz
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - David T Woodley
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
16
|
Yamaguchi Y, Nameki S, Kato Y, Saita R, Sato T, Nagao S, Murakami T, Yoshimine Y, Amiya S, Morita T, Okita Y, Kawasaki T, Fujimoto J, Ueda Y, Maeda Y, Watanabe A, Takamatsu H, Nishida S, Shima Y, Narazaki M, Kumanogoh A. Persistence of SARS-CoV-2 neutralizing antibodies and anti-Omicron IgG induced by BNT162b2 mRNA vaccine in patients with autoimmune inflammatory rheumatic disease: an explanatory study in Japan. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2022; 32:100661. [PMID: 36569794 PMCID: PMC9763057 DOI: 10.1016/j.lanwpc.2022.100661] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/02/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022]
Abstract
Background Autoimmune inflammatory rheumatic disease (AIRD) patients are at high risk of the coronavirus disease 2019 (COVID-19), but the medium-term effects of immunosuppressants on vaccine efficacy are unknown. We investigated the duration of humoral responses against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) wild-type and Omicron variant in AIRD patients administered with two doses of the BNT162b2 (Pfizer-BioNTech) vaccine. Methods Serum-neutralizing antibody (NAb) and anti-receptor-binding domain (RBD)/spike antibody levels were measured. Short- and medium-term effects of immunosuppressants were analyzed pre-vaccination (Term 1) and 14-42 days (Term 2) and 100-200 days (Term 3) after the second vaccination. Findings From Feb 1, 2021, to Feb 28, 2022, 439 AIRD patients and 146 healthy controls were investigated. The seropositivity rate and log10-NAb titers were significantly lower in AIRD patients than in controls at Terms 2 and 3. In rheumatoid arthritis patients, tumor necrosis factor-α inhibitors (TNFis) at Term 3, and older age, glucocorticoids, and abatacept at Terms 2 and 3 were risk factors for reduced responses. Anti-Omicron RBD/spike IgG levels strongly correlated with NAb titers. Interpretation Glucocorticoids, TNFis, and abatacept treatments negatively affect the longevity of humoral responses to SARS-CoV-2, including Omicron, after two vaccine doses. These findings may inform the timing of additional vaccination for AIRD patients. Funding Cloud Funding of Peace Winds Japan; Center of Innovation Program from the Ministry of Education, Culture, Sports, Science and Technology of Japan; Japan Society for the Promotion of Science KAKENHI; Japan Agency for Medical Research and Development; Kansai Economic Federation; Mitsubishi Zaidan; and Research Grant from Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology.
Collapse
Affiliation(s)
- Yuta Yamaguchi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Shinichiro Nameki
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Yasuhiro Kato
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan,Corresponding author. Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Ryotaro Saita
- Department of Medical Innovation, Osaka University Hospital, Suita, Japan
| | - Tomoharu Sato
- Department of Biostatistics and Data Science, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Sayaka Nagao
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Japan
| | - Teruaki Murakami
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Yuko Yoshimine
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Saori Amiya
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Takayoshi Morita
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Yasutaka Okita
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Takahiro Kawasaki
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Jun Fujimoto
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Yasutaka Ueda
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yuichi Maeda
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan
| | - Akane Watanabe
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Laboratory of Thermo-Therapeutics for Vascular Dysfunction, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hyota Takamatsu
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Sumiyuki Nishida
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yoshihito Shima
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Laboratory of Thermo-Therapeutics for Vascular Dysfunction, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Masashi Narazaki
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan,Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan,Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Japan,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan,Japan Agency for Medical Research and Development—Core Research for Evolutional Science and Technology (AMED–CREST), Osaka University, Osaka, Japan,Center for Advanced Modalities and DDS (CAMaD), Osaka University, Osaka, Japan
| |
Collapse
|
17
|
Bruel T, Stéfic K, Nguyen Y, Toniutti D, Staropoli I, Porrot F, Guivel-Benhassine F, Bolland WH, Planas D, Hadjadj J, Handala L, Planchais C, Prot M, Simon-Lorière E, André E, Baele G, Cuypers L, Mouthon L, Mouquet H, Buchrieser J, Sève A, Prazuck T, Maes P, Terrier B, Hocqueloux L, Schwartz O. Longitudinal analysis of serum neutralization of SARS-CoV-2 Omicron BA.2, BA.4, and BA.5 in patients receiving monoclonal antibodies. Cell Rep Med 2022; 3:100850. [PMID: 36450283 PMCID: PMC9706550 DOI: 10.1016/j.xcrm.2022.100850] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/10/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
The emergence of Omicron sublineages impacts the therapeutic efficacy of anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) monoclonal antibodies (mAbs). Here, we evaluate neutralization and antibody-dependent cellular cytotoxicity (ADCC) activities of 6 therapeutic mAbs against Delta, BA.2, BA.4, and BA.5. The Omicron subvariants escape most antibodies but remain sensitive to bebtelovimab and cilgavimab. Consistent with their shared spike sequence, BA.4 and BA.5 display identical neutralization profiles. Sotrovimab is the most efficient at eliciting ADCC. We also analyze 121 sera from 40 immunocompromised individuals up to 6 months after infusion of Ronapreve (imdevimab + casirivimab) or Evusheld (cilgavimab + tixagevimab). Sera from Ronapreve-treated individuals do not neutralize Omicron subvariants. Evusheld-treated individuals neutralize BA.2 and BA.5, but titers are reduced. A longitudinal evaluation of sera from Evusheld-treated patients reveals a slow decay of mAb levels and neutralization, which is faster against BA.5. Our data shed light on antiviral activities of therapeutic mAbs and the duration of effectiveness of Evusheld pre-exposure prophylaxis.
Collapse
Affiliation(s)
- Timothée Bruel
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France; Vaccine Research Institute, Créteil, France.
| | - Karl Stéfic
- INSERM U1259, Université de Tours, Tours, France; CHRU de Tours, National Reference Center for HIV-Associated Laboratory, Tours, France
| | - Yann Nguyen
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP.CUP, Hopital Cochin, Paris, France
| | - Donatella Toniutti
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
| | - Isabelle Staropoli
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
| | - Françoise Porrot
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
| | | | - William-Henry Bolland
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France; Université Paris Cité, École doctorale BioSPC 562, Paris, France
| | - Delphine Planas
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France; Vaccine Research Institute, Créteil, France
| | - Jérôme Hadjadj
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP.CUP, Hopital Cochin, Paris, France
| | - Lynda Handala
- INSERM U1259, Université de Tours, Tours, France; CHRU de Tours, National Reference Center for HIV-Associated Laboratory, Tours, France
| | - Cyril Planchais
- Humoral Immunology Laboratory, Institut Pasteur, Université Paris Cité, INSERM U1222, Paris, France
| | - Matthieu Prot
- G5 Evolutionary Genomics of RNA Viruses, Institut Pasteur, Université Paris Cité, Paris, France
| | - Etienne Simon-Lorière
- G5 Evolutionary Genomics of RNA Viruses, Institut Pasteur, Université Paris Cité, Paris, France
| | - Emmanuel André
- University Hospitals Leuven, Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, Leuven, Belgium; KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, Leuven, Belgium
| | - Guy Baele
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical and Epidemiological Virology, Leuven, Belgium
| | - Lize Cuypers
- University Hospitals Leuven, Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, Leuven, Belgium
| | - Luc Mouthon
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP.CUP, Hopital Cochin, Paris, France
| | - Hugo Mouquet
- Humoral Immunology Laboratory, Institut Pasteur, Université Paris Cité, INSERM U1222, Paris, France
| | - Julian Buchrieser
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
| | - Aymeric Sève
- CHR d'Orléans, Service de Maladies Infectieuses, Orléans, France
| | - Thierry Prazuck
- CHR d'Orléans, Service de Maladies Infectieuses, Orléans, France
| | - Piet Maes
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical and Epidemiological Virology, Leuven, Belgium
| | - Benjamin Terrier
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP.CUP, Hopital Cochin, Paris, France
| | | | - Olivier Schwartz
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France; Vaccine Research Institute, Créteil, France.
| |
Collapse
|
18
|
Hosseini SSJ, Dudakova A, Kummer K, Zschüntzsch J. [SARS-CoV-2 antibody response to the second COVID-19 vaccination in neuromuscular disease patients under immune modulating treatment]. DER NERVENARZT 2022; 93:1219-1227. [PMID: 35997783 PMCID: PMC9395911 DOI: 10.1007/s00115-022-01363-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 12/04/2022]
Abstract
Successful vaccination (adequate elevation of anti-spike protein antibodies) is attributed with sufficient protection against a severe course of coronavirus disease 2019 (COVID-19). For patients with chronic inflammatory diseases (CID) and immunosuppression the success of vaccination is an ongoing scientific discourse. Therefore, we evaluated the antibody titer against the S1 antigen of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) 2 weeks after complete immunization in patients with an underlying neuromuscular disease (NMD), who presented to our neurological day clinic and outpatient department for regular infusions of immunoglobulins. The data show that patients with chronic autoimmune NMD and simultaneous immunosuppressive or immune modulating treatment show an antibody response after vaccination with both mRNA and vector vaccines. In comparison to healthy subjects there is a comparable number of seroconversions due to the vaccination. A correlation between immunoglobulin dose and vaccination response could not be found; however, in contrast, there was a significant reduction of specific antibody synthesis, especially for the combination of mycophenolate mofetil (MMF) and prednisolone.
Collapse
Affiliation(s)
- S S Justus Hosseini
- Neuromuskuläres Zentrum Göttingen, Klinik für Neurologie, Universitätsmedizin Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Deutschland
| | - Anna Dudakova
- Institut für Medizinische Mikrobiologie und Virologie, Universitätsmedizin Göttingen, Göttingen, Deutschland
| | - Karsten Kummer
- Neuromuskuläres Zentrum Göttingen, Klinik für Neurologie, Universitätsmedizin Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Deutschland
| | - Jana Zschüntzsch
- Neuromuskuläres Zentrum Göttingen, Klinik für Neurologie, Universitätsmedizin Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Deutschland.
| |
Collapse
|
19
|
Sánchez-Zapardiel E, Alós M, Nozal P, González-Muñoz M, Frauca-Remacha E, Gavilán LB, Quiles MJ, Hierro L, López-Granados E. Humoral and cellular immune responses to Pfizer-BioNTech BNT162b2 SARS-CoV-2 vaccine in adolescents with liver transplantation: Single center experience. Front Immunol 2022; 13:1049188. [PMID: 36505469 PMCID: PMC9727154 DOI: 10.3389/fimmu.2022.1049188] [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: 09/20/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
Background Immune responses to vaccines against severe acute respiratory syndrome (SARS)-coronavirus (CoV)-2 are variable. In the absence of disease, youngsters are expected to better react to vaccines than adults. Nevertheless, chronic immunosuppression in transplant recipients may impair their capability to generate protection. We aim to explore immune responses after BNT162b2 SARS-CoV-2 vaccination in our cohort of young liver-transplanted patients. Methods A prospective study of adolescent liver-transplanted patients (n=33) in the long-term follow-up was performed. Immune responses after receiving Pfizer-BioNTech BNT162b2 vaccine were analyzed at two time-points: baseline and 30 days after the second dose. Humoral responses were measured by fluoroenzyme-immunoassay and T-cell responses by interferon-γ-release assay. Post-vaccine coronavirus disease (COVID-19) events were recorded by a survey. Results Pre-vaccine SARS-CoV-2-specific antibodies were undetectable in 27/32 (84.4%), negative/indeterminate in 3/32 (9.4%) and positive in 2/32 (6.3%) patients. Cellular responses at baseline were negative in 12/18 (66.6%), positive in 3/18 (16.6%) and indeterminate in 3/18 (16.6%) recipients. None of the baseline positives recalled any symptoms. Post-vaccine antibodies were detected in all patients and 92.6% showed levels >816 BAU/mL. Twenty (71.4%) recipients had positive T-cell responses. Regarding post-vaccine SARS-Cov-2 infection, 10 (30.3%) patients reported COVID-19 without hospitalization and 21 (63.6%) did not notify any infection. Negative and positive cell-response groups after vaccination showed statistically significant differences regarding COVID-19 cases (62.5% vs 22.2%, respectively; p=0.046). Conclusions Adolescents and young adults with liver transplantation responded to SARS-Cov-2 vaccine, generating both humoral and cellular responses. Recipients developing cellular responses after vaccination had a lower incidence of COVID-19.
Collapse
Affiliation(s)
- Elena Sánchez-Zapardiel
- Department of Immunology, La Paz University Hospital, Madrid, Spain,Lymphocyte Pathophysiology in Immunodeficiency Group, La Paz Biomedical Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain,European Reference Network (ERN) Transplant-Child, Madrid, Spain,*Correspondence: Elena Sánchez-Zapardiel,
| | - María Alós
- European Reference Network (ERN) Transplant-Child, Madrid, Spain,Department of Pediatric Hepatology, La Paz University Hospital, Madrid, Spain
| | - Pilar Nozal
- Department of Immunology, La Paz University Hospital, Madrid, Spain,Diagnosis and Treatment of Pathologies Associated with Alterations of the Complement System Group, La Paz Biomedical Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain,Rare Diseases Networking Biomedical Research Centre (CIBERER U754), Madrid, Spain
| | - Miguel González-Muñoz
- Department of Immunology, La Paz University Hospital, Madrid, Spain,Patient Safety and Quality Research Group, La Paz Biomedical Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain
| | - Esteban Frauca-Remacha
- European Reference Network (ERN) Transplant-Child, Madrid, Spain,Department of Pediatric Hepatology, La Paz University Hospital, Madrid, Spain
| | | | - María José Quiles
- Department of Pediatric Hepatology, La Paz University Hospital, Madrid, Spain
| | - Loreto Hierro
- European Reference Network (ERN) Transplant-Child, Madrid, Spain,Department of Pediatric Hepatology, La Paz University Hospital, Madrid, Spain
| | - Eduardo López-Granados
- Department of Immunology, La Paz University Hospital, Madrid, Spain,Lymphocyte Pathophysiology in Immunodeficiency Group, La Paz Biomedical Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain,European Reference Network (ERN) Transplant-Child, Madrid, Spain,Rare Diseases Networking Biomedical Research Centre (CIBERER U767), Madrid, Spain
| |
Collapse
|
20
|
Furer V, Eviatar T, Freund T, Peleg H, Paran D, Levartovsky D, Kaufman I, Broyde A, Elalouf O, Polachek A, Feld J, Haddad A, Gazitt T, Elias M, Higazi N, Kharouf F, Gertel S, Pel S, Nevo S, Hagin D, Zisman D, Elkayam O. Immunogenicity induced by two and three doses of the BNT162b2 mRNA vaccine in patients with autoimmune inflammatory rheumatic diseases and immunocompetent controls: a longitudinal multicentre study. Ann Rheum Dis 2022; 81:1594-1602. [PMID: 35868846 DOI: 10.1136/ard-2022-222550] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 07/02/2022] [Indexed: 12/27/2022]
Abstract
OBJECTIVES To evaluate long-term kinetics of the BNT162b2 mRNA vaccine-induced immune response in adult patients with autoimmune inflammatory rheumatic diseases (AIIRD) and immunocompetent controls. METHODS A prospective multicentre study investigated serum anti-SARS-CoV-2 S1/S2 IgG titre at 2-6 weeks (AIIRD n=720, controls n=122) and 6 months (AIIRD n=628, controls n=116) after the second vaccine, and 2-6 weeks after the third vaccine dose (AIIRD n=169, controls n=45). T-cell immune response to the third vaccine was evaluated in a small sample. RESULTS The two-dose vaccine regimen induced a higher humoral response in controls compared with patients, postvaccination seropositivity rates of 100% versus 84.72%, p<0.0001, and 96.55% versus 74.26%, p<0.0001 at 2-6 weeks and at 6 months, respectively. The third vaccine dose restored the seropositive response in all controls and 80.47% of patients with AIIRD, p=0.0028. All patients treated with methotrexate monotherapy, anticytokine biologics, abatacept and janus kinase (JAK) inhibitors regained the humoral response after the third vaccine, compared with only a third of patients treated with rituximab, entailing a 16.1-fold risk for a negative humoral response, p≤0.0001. Cellular immune response in rituximab-treated patients was preserved before and after the third vaccine and was similar to controls. Breakthrough COVID-19 rate during the Delta surge was similar in patients and controls, 1.83% versus 1.43%, p=1. CONCLUSIONS The two-dose BNTb262 regimen was associated with similar clinical efficacy and similar waning of the humoral response over 6 months among patients with AIIRD and controls. The third vaccine dose restored the humoral response in all of the controls and the majority of patients.
Collapse
Affiliation(s)
- Victoria Furer
- Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Tali Eviatar
- Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Tal Freund
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Hagit Peleg
- Rheumatology, Hadassah University Medical Center, Jerusalem, Israel
| | - Daphna Paran
- Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | | | - Ilana Kaufman
- Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Adi Broyde
- Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ofir Elalouf
- Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Ari Polachek
- Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Joy Feld
- Carmel Medical Center, Haifa, Israel
- Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Amir Haddad
- Carmel Medical Center, Haifa, Israel
- Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Tal Gazitt
- Carmel Medical Center, Haifa, Israel
- Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Muna Elias
- Carmel Medical Center, Haifa, Israel
- Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Nizar Higazi
- Carmel Medical Center, Haifa, Israel
- Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Fadi Kharouf
- Rheumatology, Hadassah University Medical Center, Jerusalem, Israel
| | - Smadar Gertel
- Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Sara Pel
- Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Sharon Nevo
- Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - David Hagin
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Devy Zisman
- Carmel Medical Center, Haifa, Israel
- Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Ori Elkayam
- Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| |
Collapse
|
21
|
Zhao J, Zhu J, Huang C, Zhu X, Zhu Z, Wu Q, Yuan R. Uncovering the information immunology journals transmitted for COVID-19: A bibliometric and visualization analysis. Front Immunol 2022; 13:1035151. [PMID: 36405695 PMCID: PMC9670819 DOI: 10.3389/fimmu.2022.1035151] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/17/2022] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Since the global epidemic of the coronavirus disease 2019 (COVID-19), a large number of immunological studies related to COVID-19 have been published in various immunology journals. However, the results from these studies were discrete, and no study summarized the important immunological information about COVID-19 released by these immunology journals. This study aimed to comprehensively summarize the knowledge structure and research hotspots of COVID-19 published in major immunology journals through bibliometrics. METHODS Publications on COVID-19 in major immunology journals were obtained from the Web of Science Core Collection. CiteSpace, VOSviewer, and R-bibliometrix were comprehensively used for bibliometric and visual analysis. RESULTS 1,331 and 5,000 publications of 10 journals with high impact factors and 10 journals with the most papers were included, respectively. The USA, China, England, and Italy made the most significant contributions to these papers. University College London, National Institute of Allergy and Infectious Diseases, Harvard Medical School, University California San Diego, and University of Pennsylvania played a central role in international cooperation in the immunology research field of COVID-19. Yuen Kwok Yung was the most important author in terms of the number of publications and citations, and the H-index. CLINICAL INFECTIOUS DISEASES and FRONTIERS IN IMMUNOLOGY were the most essential immunology journals. These immunology journals mostly focused on the following topics: "Delta/Omicron variants", "cytokine storm", "neutralization/neutralizing antibody", "T cell", "BNT162b2", "mRNA vaccine", "vaccine effectiveness/safety", and "long COVID". CONCLUSION This study systematically uncovered a holistic picture of the current research on COVID-19 published in major immunology journals from the perspective of bibliometrics, which will provide a reference for future research in this field.
Collapse
Affiliation(s)
- Jiefeng Zhao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jinfeng Zhu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Chao Huang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaojian Zhu
- Center for Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhengming Zhu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qinrong Wu
- Department of General Surgery, Yingtan City People’s Hospital, Yingtan, Jiangxi, China
| | - Rongfa Yuan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
22
|
Patients with Hematological Malignancies Treated with T-Cell or B-Cell Immunotherapy Remain at High Risk of Severe Forms of COVID-19 in the Omicron Era. Viruses 2022; 14:v14112377. [PMID: 36366475 PMCID: PMC9696091 DOI: 10.3390/v14112377] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Patients with hematological malignancies are at greater risk of severe COVID-19 and have been prioritized for COVID-19 vaccination. A significant proportion of them have an impaired vaccine response, both due to the underlying disease and to the treatments. METHODS We conducted a prospective observational study to identify the specific risks of the outpatient population with hematological diseases. RESULT Between 22 December 2021 to 12 February 2022, we followed 338 patients of which 16.9% (n = 57) developed SARS-CoV-2 infection despite previous vaccination (94.7%). COVID-19 patients were more likely to have received immunotherapy (85.5% vs. 41%, p < 10-4), and particularly anti-CD20 monoclonal antibodies (40% vs. 14.9%, p < 10-4) and Bruton's tyrosine kinase inhibitors (BTKi) (7.3% vs. 0.7%, p < 10-2). There was no significant difference in demographic characteristics or hematological malignancies between COVID-19-positive and non-positive patients. Patients hospitalized for COVID-19 had more frequently received immunotherapy than patients with asymptomatic or benign forms (100% vs. 77.3%, p < 0.05). Hospitalized COVID-19 patients had a higher proportion of negative or weakly positive serologies than non-hospitalized patients (92.3% vs. 61%, p < 0.05). Patients who received tixagevimab/cilgavimab prophylaxis (n = 102) were less likely to be COVID-19-positive (4.9 vs. 22%, p < 0.05) without significant difference in hospitalization rates. CONCLUSION In the immunocompromised population of patients with hematological malignancies, the underlying treatment of blood cancer by immunotherapy appears to be a risk factor for SARS-CoV-2 infection and for developing a severe form.
Collapse
|
23
|
Bruel T, Pinaud L, Tondeur L, Planas D, Staropoli I, Porrot F, Guivel-Benhassine F, Attia M, Pelleau S, Woudenberg T, Duru C, Koffi AD, Castelain S, Fernandes-Pellerin S, Jolly N, De Facci LP, Roux E, Ungeheuer MN, Van Der Werf S, White M, Schwartz O, Fontanet A. Neutralising antibody responses to SARS-CoV-2 omicron among elderly nursing home residents following a booster dose of BNT162b2 vaccine: A community-based, prospective, longitudinal cohort study. EClinicalMedicine 2022; 51:101576. [PMID: 35891947 PMCID: PMC9307278 DOI: 10.1016/j.eclinm.2022.101576] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The protective immunity against omicron following a BNT162b2 Pfizer booster dose among elderly individuals (ie, those aged >65 years) is not well characterised. METHODS In a community-based, prospective, longitudinal cohort study taking place in France in which 75 residents from three nursing homes were enrolled, we selected 38 residents who had received a two-dose regimen of mRNA vaccine and a booster dose of Pfizer BNT162b2 vaccine. We excluded individuals that did not receive three vaccine doses or did not have available sera samples. We measured anti-S IgG antibodies and neutralisation capacity in sera taken 56 (28-68) and 55 (48-64) days (median (range)) after the 2nd and 3rd vaccine doses, respectively. Antibodies targeting the SARS-CoV-2 Spike protein were measured with the S-Flow assay as binding antibody units per milliliter (BAU/mL). Neutralising activities in sera were measured as effective dilution 50% (ED50) with the S-Fuse assay using authentic isolates of delta and omicron BA.1. FINDINGS Among the 38 elderly individuals recruited to the cohort study between November 23rd, 2020 and April 29th, 2021, with median age of 88 (range 72-101) years, 30 (78.95%) had been previously infected with SARS-CoV-2. After three vaccine doses, serum neutralising activity was lower against omicron BA.1 (median ED50 of 774.5, range 15.0-34660.0) than the delta variant (median ED50 of 4972.0, range 213.7-66340.0), and higher among previously infected (ie, convalescent; median ED50 against omicron: 1088.0, range 32.6-34660.0) compared with infection-naive residents (median ED50 against omicron: 188.4, range 15.0-8918.0). During the French omicron wave in December 2021-January 2022, 75% (6/8) of naive residents were infected, compared to 25% (7/30) of convalescent residents (P=0.0114). Anti-Spike antibody levels and neutralising activity against omicron BA.1 after a third BNT162b2 booster dose were lower in those with breakthrough BA.1 infection (n=13) compared with those without (n=25), with a median of 1429.9 (range 670.9-3818.3) BAU/mL vs 2528.3 (range 695.4-8832.0) BAU/mL (P=0.029) and a median ED50 of 281.1 (range 15.0-2136.0) vs 1376.0 (range 32.6-34660.0) (P=0.0013), respectively. INTERPRETATION This study shows that elderly individuals who received three vaccine doses elicit neutralising antibodies against the omicron BA.1 variant of SARS-CoV-2. Elderly individuals who had also been previously infected showed higher neutralising activity compared with naive individuals. Yet, breakthrough infections with omicron occurred. Individuals with breakthrough infections had significantly lower neutralising titers compared to individuals without breakthrough infection. Thus, a fourth dose of vaccine may be useful in the elderly population to increase the level of neutralising antibodies and compensate for waning immunity. FUNDING Institut Pasteur, Fondation pour la Recherche Médicale (FRM), European Health Emergency Preparedness and Response Authority (HERA), Agence nationale de recherches sur le sida et les hépatites virales - Maladies Infectieuses Emergentes (ANRS-MIE), Agence nationale de la recherche (ANR), Assistance Publique des Hôpitaux de Paris (AP-HP) and Fondation de France.
Collapse
Affiliation(s)
- Timothée Bruel
- Virus & Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
- Vaccine Research Institute, Créteil, France
- Corresponding author at: Unité Virus et Immunité, Institut Pasteur, 25-28 Rue du docteur Roux, 75015 Paris, France.
| | - Laurie Pinaud
- Emerging Diseases Epidemiology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Laura Tondeur
- Emerging Diseases Epidemiology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Delphine Planas
- Virus & Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
- Vaccine Research Institute, Créteil, France
| | - Isabelle Staropoli
- Virus & Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
| | - Françoise Porrot
- Virus & Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
| | | | - Mikaël Attia
- Molecular Genetics of RNA Viruses Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Stéphane Pelleau
- Infectious Disease Epidemiology and Analytics Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Tom Woudenberg
- Infectious Disease Epidemiology and Analytics Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Cécile Duru
- Hôpital de Crépy-en-Valois, Crépy-en-Valois, France
| | | | | | | | - Nathalie Jolly
- Centre for Translational Science, Institut Pasteur, Paris, France
| | - Louise Perrin De Facci
- Clinical Investigation and access to bioresources (ICAReB) platform, Centre for Translational Science, Institut Pasteur, Paris, France
| | - Emmanuel Roux
- Clinical Investigation and access to bioresources (ICAReB) platform, Centre for Translational Science, Institut Pasteur, Paris, France
| | - Marie-Noëlle Ungeheuer
- Clinical Investigation and access to bioresources (ICAReB) platform, Centre for Translational Science, Institut Pasteur, Paris, France
| | - Sylvie Van Der Werf
- Molecular Genetics of RNA Viruses Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Michael White
- Infectious Disease Epidemiology and Analytics Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Olivier Schwartz
- Virus & Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
- Vaccine Research Institute, Créteil, France
| | - Arnaud Fontanet
- Emerging Diseases Epidemiology Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Conservatoire National des Arts et Métiers, PACRI Unit, Paris, France
- Corresponding author at: Emerging Diseases Epidemiology Unit, Institut Pasteur, 25-28 Rue du docteur Roux, 75015 Paris, France.
| |
Collapse
|
24
|
Lafont E, Pere H, Lebeaux D, Cheminet G, Thervet E, Guillemain R, Flahault A. Targeted SARS-CoV-2 treatment is associated with decreased mortality in immunocompromised patients with COVID-19. J Antimicrob Chemother 2022; 77:2688-2692. [PMID: 35876174 PMCID: PMC9384481 DOI: 10.1093/jac/dkac253] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/06/2022] [Indexed: 12/29/2022] Open
Abstract
Background Little is known about targeted (antiviral or monoclonal antibody) anti-SARS-CoV-2 treatment in immunocompromised patients with COVID-19. Objectives To assess the real-life efficacy and tolerance of targeted treatment of COVID-19 in immunocompromised patients. Patients and methods Single-centre retrospective case series of immunocompromised patients with COVID-19 between December 2021 and March 2022. We recorded all cases of COVID-19 among immunocompromised patients treatment between 20 December 2021 and 15 March 2022. Choice of treatment was left to the physician’s decision, according to internal treatment protocol, treatment availability and circulating variants. Main outcome was death from COVID-19 after no treatment or targeted treatment. Results Sixty-seven immunocompromised patients [38 male; median (IQR) age, 53 (43–63) years], with a median (IQR) follow-up of 60 (47–80) days. Ten patients did not receive any targeted treatment. Targeted treatment consisted of IV curative remdesivir (n = 22), sotrovimab (n = 16), tixagevimab/cilgavimab (n = 13) and casirivimab/imdevimab (n = 1). Ten patients (15%) presented severe COVID-19 and 2 (3%) died from Omicron COVID-19. Comparing patients who received targeted anti-SARS-CoV-2 treatment and no prophylaxis, (n = 42; 81%) with those who did not (n = 10; 19%), death rate was significantly lower in treated patients [n = 0 (0%) versus n = 2 (20%); P = 0.034]. No severe adverse events were reported among treated patients. Among 15 patients who received tixagevimab/cilgavimab as pre-exposure prophylaxis, 6 received an additional curative treatment and none died from COVID-19. Conclusions Our results suggest that targeted COVID-19 treatment, including direct antivirals or monoclonal antibodies, is safe and efficient and could be proposed in high-risk immunocompromised patients.
Collapse
Affiliation(s)
- Emmanuel Lafont
- Service de Médecine Interne, Hôpital Européen Georges Pompidou, AP-HP Centre, Université Paris Cité, Paris, France.,Université Paris Cité, Paris, France
| | - Hélène Pere
- Université Paris Cité, Paris, France.,Service de Microbiologie, Unité de Virologie, Hôpital Européen Georges Pompidou, AP-HP Centre, Université Paris Cité, Paris, France
| | - David Lebeaux
- Université Paris Cité, Paris, France.,Service de Microbiologie, Unité Mobile d'Infectiologie, Hôpital Européen Georges Pompidou, AP-HP Centre, Université Paris Cité, Paris, France
| | - Geoffrey Cheminet
- Service de Médecine Interne, Hôpital Européen Georges Pompidou, AP-HP Centre, Université Paris Cité, Paris, France.,Université Paris Cité, Paris, France
| | - Eric Thervet
- Université Paris Cité, Paris, France.,Service de Néphrologie, Hôpital Européen Georges Pompidou, AP-HP Centre, Université Paris Cité, Paris, France
| | - Romain Guillemain
- Service de Chirurgie Cardiaque, Hôpital Européen Georges Pompidou, AP-HP. Centre, Université de Paris Cité, Paris, France
| | - Adrien Flahault
- Université Paris Cité, Paris, France.,Service de Néphrologie, Hôpital Européen Georges Pompidou, AP-HP Centre, Université Paris Cité, Paris, France
| |
Collapse
|
25
|
Hirai T, Yoshioka Y. Considerations of CD8+ T Cells for Optimized Vaccine Strategies Against Respiratory Viruses. Front Immunol 2022; 13:918611. [PMID: 35774782 PMCID: PMC9237416 DOI: 10.3389/fimmu.2022.918611] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
The primary goal of vaccines that protect against respiratory viruses appears to be the induction of neutralizing antibodies for a long period. Although this goal need not be changed, recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have drawn strong attention to another arm of acquired immunity, CD8+ T cells, which are also called killer T cells. Recent evidence accumulated during the coronavirus disease 2019 (COVID-19) pandemic has revealed that even variants of SARS-CoV-2 that escaped from neutralizing-antibodies that were induced by either infection or vaccination could not escape from CD8+ T cell-mediated immunity. In addition, although traditional vaccine platforms, such as inactivated virus and subunit vaccines, are less efficient in inducing CD8+ T cells, newly introduced platforms for SARS-CoV-2, namely, mRNA and adenoviral vector vaccines, can induce strong CD8+ T cell-mediated immunity in addition to inducing neutralizing antibodies. However, CD8+ T cells function locally and need to be at the site of infection to control it. To fully utilize the protective performance of CD8+ T cells, it would be insufficient to induce only memory cells circulating in blood, using injectable vaccines; mucosal immunization could be required to set up CD8+ T cells for the optimal protection. CD8+ T cells might also contribute to the pathology of the infection, change their function with age and respond differently to booster vaccines in comparison with antibodies. Herein, we overview cutting-edge ideas on CD8+ T cell-mediated immunity that can enable the rational design of vaccines for respiratory viruses.
Collapse
Affiliation(s)
- Toshiro Hirai
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- *Correspondence: Toshiro Hirai,
| | - Yasuo Yoshioka
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- The Research Foundation for Microbial Diseases of Osaka University, Suita, Japan
| |
Collapse
|
26
|
Design, immunogenicity, and efficacy of a pan-sarbecovirus dendritic-cell targeting vaccine. EBioMedicine 2022; 80:104062. [PMID: 35594660 PMCID: PMC9113741 DOI: 10.1016/j.ebiom.2022.104062] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/11/2022] [Accepted: 04/29/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND There is an urgent need of a new generation of vaccine that are able to enhance protection against SARS-CoV-2 and related variants of concern (VOC) and emerging coronaviruses. METHODS We identified conserved T- and B-cell epitopes from Spike (S) and Nucleocapsid (N) highly homologous to 38 sarbecoviruses, including SARS-CoV-2 VOCs, to design a protein subunit vaccine targeting antigens to Dendritic Cells (DC) via CD40 surface receptor (CD40.CoV2). FINDINGS CD40.CoV2 immunization elicited high levels of cross-neutralizing antibodies against SARS-CoV-2, VOCs, and SARS-CoV-1 in K18-hACE2 transgenic mice, associated with viral control and survival after SARS-CoV-2 challenge. A direct comparison of CD40.CoV2 with the mRNA BNT162b2 vaccine showed that the two vaccines were equally immunogenic in mice. We demonstrated the potency of CD40.CoV2 to recall in vitro human multi-epitope, functional, and cytotoxic SARS-CoV-2 S- and N-specific T-cell responses that are unaffected by VOC mutations and cross-reactive with SARS-CoV-1 and, to a lesser extent, MERS epitopes. INTERPRETATION We report the immunogenicity and antiviral efficacy of the CD40.CoV2 vaccine in a preclinical model providing a framework for a pan-sarbecovirus vaccine. FUNDINGS This work was supported by INSERM and the Investissements d'Avenir program, Vaccine Research Institute (VRI), managed by the ANR and the CARE project funded from the Innovative Medicines Initiative 2 Joint Undertaking (JU).
Collapse
|
27
|
Assawasaksakul T, Lertussavavivat T, Sathitratanacheewin S, Oudomying N, Vichaiwattana P, Wanlapakorn N, Poovorawan Y, Avihingsanon Y, Assawasaksakul N, Buranapraditkun S, Kittanamongkolchai W. Comparison of Immunogenicity and Safety of Inactivated, Adenovirus-Vectored, and Heterologous Adenovirus-Vectored/mRNA Vaccines in Patients with Systemic Lupus Erythematosus and Rheumatoid Arthritis: A Prospective Cohort Study. Vaccines (Basel) 2022; 10:vaccines10060853. [PMID: 35746461 PMCID: PMC9227480 DOI: 10.3390/vaccines10060853] [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] [Received: 04/25/2022] [Revised: 05/22/2022] [Accepted: 05/24/2022] [Indexed: 01/02/2023] Open
Abstract
Background: Impaired immune responses to COVID-19 vaccines have been observed in autoimmune rheumatic disease patients. Determining the most effective and safe vaccine regimen is critically needed in such a population. We aim to compare the immunogenicity and safety of three COVID-19 vaccine regimens in patients with systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Methods: SLE and RA patients aged 18−65 years who received inactivated (CoronaVac or COVILO), adenovirus-vectored (AZD1222), or heterogeneous (AZD1222/BNT162b2) vaccines were enrolled. Humoral and cellular immune responses were assessed at day 28 after the second vaccination. This was performed using the serum binding antibody level against the receptor-binding domain of the SARS-CoV-2 spike protein (anti-RBD Ig) and IFNy-ELISpot assay (ELISpot), respectively. Reactogenicity was reviewed on day 7 following each vaccination. Disease activity was assessed before and on day 28 after the second vaccination. Results: The cohort consisted of 94 patients (64 SLE and 30 RA). Inactivated, AZD1222, and AZD1222/BNT162b2 vaccines were administered to 23, 43, and 28 patients, respectively. Anti-RBD titers were lowest in the inactivated vaccine group (2.84 AU/mL; 95% CI 0.96−8.44), followed by AZD1222 (233.7 AU/mL; 95% CI 99.0−505.5), and AZD1222/BNT162b2 (688.6 AU/mL; 95% CI 271−1745), p < 0.0001. After adjusting for relevant factors, the inactivated vaccine was associated with the lowest humoral response, while adenovirus-vectored/mRNA vaccine was the highest. The proportion of positive ELISpot test was also lowest in the inactivated vaccine group (27%), followed by the adenovirus-vectored vaccine (67%), and the adenovirus-vectored/mRNA vaccine (73%) (p = 0.03). All types of vaccine were well-tolerated. There was no flare of autoimmune disease post-vaccination. Conclusion: Adenovirus-vectored and adenovirus-vectored/mRNA vaccines elicited a stronger humoral and cellular immune response than inactivated vaccines, suggesting that they may be more suitable in SLE and RA patients receiving immunosuppressive therapy.
Collapse
Affiliation(s)
- Theerada Assawasaksakul
- Division of Rheumatology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Tanat Lertussavavivat
- Division of Nephrology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (T.L.); (Y.A.)
| | - Seelwan Sathitratanacheewin
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.); (N.O.); (S.B.)
| | - Nont Oudomying
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.); (N.O.); (S.B.)
| | - Preeyaporn Vichaiwattana
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.V.); (N.W.); (Y.P.)
| | - Nasamon Wanlapakorn
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.V.); (N.W.); (Y.P.)
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.V.); (N.W.); (Y.P.)
- The Royal Society of Thailand, Sanam Sueapa, Dusit, Bangkok 10300, Thailand
| | - Yingyos Avihingsanon
- Division of Nephrology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (T.L.); (Y.A.)
| | | | - Supranee Buranapraditkun
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.); (N.O.); (S.B.)
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wonngarm Kittanamongkolchai
- Division of Nephrology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (T.L.); (Y.A.)
- Mahachakri Sirindhorn Clinical Research Center, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Renal Immunology and Transplantation Research Unit, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Rd., Pathumwan, Bangkok 10330, Thailand
- Correspondence:
| |
Collapse
|
28
|
Liew J, Gianfrancesco M, Harrison C, Izadi Z, Rush S, Lawson-Tovey S, Jacobsohn L, Ja C, Hyrich KL, Gossec L, Strangfeld A, Carmona L, Schäfer M, Frãzao-Mateus E, Bulina I, Stafford F, Tufan A, Graver C, Yardımcı GK, Zepa J, Al Emadi S, Cook C, Abutiban F, Dey D, Katigbak G, Kaufman L, Kowalski E, Martínez-Martínez MU, Patel NJ, Reyes-Cordero G, Salido E, Smith E, Snow D, Sparks J, Wise L, Bhana S, Gore-Massy M, Grainger R, Hausmann J, Sirotich E, Sufka P, Wallace Z, Machado PM, Robinson PC, Yazdany J. SARS-CoV-2 breakthrough infections among vaccinated individuals with rheumatic disease: results from the COVID-19 Global Rheumatology Alliance provider registry. RMD Open 2022; 8:e002187. [PMID: 35387864 PMCID: PMC8987210 DOI: 10.1136/rmdopen-2021-002187] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/09/2022] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE While COVID-19 vaccination prevents severe infections, poor immunogenicity in immunocompromised people threatens vaccine effectiveness. We analysed the clinical characteristics of patients with rheumatic disease who developed breakthrough COVID-19 after vaccination against SARS-CoV-2. METHODS We included people partially or fully vaccinated against SARS-CoV-2 who developed COVID-19 between 5 January and 30 September 2021 and were reported to the Global Rheumatology Alliance registry. Breakthrough infections were defined as occurring ≥14 days after completion of the vaccination series, specifically 14 days after the second dose in a two-dose series or 14 days after a single-dose vaccine. We analysed patients' demographic and clinical characteristics and COVID-19 symptoms and outcomes. RESULTS SARS-CoV-2 infection was reported in 197 partially or fully vaccinated people with rheumatic disease (mean age 54 years, 77% female, 56% white). The majority (n=140/197, 71%) received messenger RNA vaccines. Among the fully vaccinated (n=87), infection occurred a mean of 112 (±60) days after the second vaccine dose. Among those fully vaccinated and hospitalised (n=22, age range 36-83 years), nine had used B cell-depleting therapy (BCDT), with six as monotherapy, at the time of vaccination. Three were on mycophenolate. The majority (n=14/22, 64%) were not taking systemic glucocorticoids. Eight patients had pre-existing lung disease and five patients died. CONCLUSION More than half of fully vaccinated individuals with breakthrough infections requiring hospitalisation were on BCDT or mycophenolate. Further risk mitigation strategies are likely needed to protect this selected high-risk population.
Collapse
Affiliation(s)
- Jean Liew
- Medicine, Section of Rheumatology, Boston University, Boston, Massachusetts, USA
| | - Milena Gianfrancesco
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, California, USA
| | | | - Zara Izadi
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
| | - Stephanie Rush
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Saskia Lawson-Tovey
- Centre for Genetics and Genomics Versus Arthritis, The University of Manchester, Manchester, UK
- National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Lindsay Jacobsohn
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, California, USA
| | - Clairissa Ja
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, California, USA
| | | | - Laure Gossec
- INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, Sorbonne Universite, Paris, France
- APHP, Rheumatology Department, Hopital Universitaire Pitie Salpetriere, Paris, France
| | - Anja Strangfeld
- Forschungsbereich Epidemiologie, Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany
| | - Loreto Carmona
- Instituto de Salud Musculoesquelética (INMUSC), Madrid, Spain
| | - Martin Schäfer
- Epidemiology and Health Services Research, German Rheumatism Research Center Berlin, Berlin, Germany
| | | | - Inita Bulina
- Paul Stradins Clinical University Hospital, Riga, Latvia
| | | | | | | | - Gözde Kübra Yardımcı
- Department of Internal Medicine, Hacettepe University, Ankara, Turkey
- Hacettepe University, Ankara, Turkey
| | - Julija Zepa
- Paul Stradins Clinical University Hospital, Riga, Latvia
| | | | - Claire Cook
- Rheumatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Dfiza Dey
- University of Ghana Medical School, Accra, Ghana
- Korle Bu Teaching Hospital, Accra, Ghana
| | | | - Lauren Kaufman
- Rheumatology Associates Louisville, Louisville, Kentucky, USA
| | - Emily Kowalski
- Inflammation and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Marco Ulises Martínez-Martínez
- Rheumatology, Hospital Central "Dr Ignacio Morones Prieto", San Luis Potosí, Mexico
- Faculty of Medicine, Universidad Autónoma de San Luis Potosí, San Luis, Mexico
| | - Naomi J Patel
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Evelyn Salido
- University of the Philippines Manila, Manila, Philippines
| | - Ellison Smith
- University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
- Asheville Arthritis & Osteoporosis Center, Asheville, North Carolina, USA
| | - David Snow
- Cape Fear Arthritis Care, Leland, North Carolina, USA
| | - Jeffrey Sparks
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Leanna Wise
- Department of Internal Medicine, Division of Rheumatology, University of Southern California, Los Angeles, California, USA
| | | | | | - Rebecca Grainger
- Department of Medicine, University of Otago, Wellington, Wellington, New Zealand
- University Of Otago, Wellington, New Zealand
| | - Jonathan Hausmann
- Rheumatology, Boston Children's Hospital, Boston, Massachusetts, USA
- Division of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Emily Sirotich
- Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | - Zachary Wallace
- Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Pedro M Machado
- MRC Centre for Neuromuscular Diseases, University College London, London, UK
- Rheumatology, University College London Centre for Rheumatology, London, UK
| | - Philip C Robinson
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
- Metro North Hospital and Health Service, Royal Brisbane and Women's Hospital Health Service District, Herston, Queensland, Australia
| | - Jinoos Yazdany
- Medicine/Rheumatology, University of California, San Francisco, California, USA
| |
Collapse
|
29
|
Bruel T, Hadjadj J, Maes P, Planas D, Seve A, Staropoli I, Guivel-Benhassine F, Porrot F, Bolland WH, Nguyen Y, Casadevall M, Charre C, Péré H, Veyer D, Prot M, Baidaliuk A, Cuypers L, Planchais C, Mouquet H, Baele G, Mouthon L, Hocqueloux L, Simon-Loriere E, André E, Terrier B, Prazuck T, Schwartz O. Serum neutralization of SARS-CoV-2 Omicron sublineages BA.1 and BA.2 in patients receiving monoclonal antibodies. Nat Med 2022; 28:1297-1302. [PMID: 35322239 DOI: 10.1038/s41591-022-01792-5] [Citation(s) in RCA: 210] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 03/22/2022] [Indexed: 11/09/2022]
Abstract
The SARS-CoV-2 Omicron BA.1 sublineage has been supplanted in many countries by the BA.2 sublineage. BA.2 differs from BA.1 by about 21 mutations in its spike. Here, we first compared the sensitivity of BA.1 and BA.2 to neutralization by 9 therapeutic monoclonal antibodies (mAbs). In contrast to BA.1, BA.2 was sensitive to Cilgavimab, partly inhibited by Imdevimab and resistant to Adintrevimab and Sotrovimab. We then analyzed sera from 29 immunocompromised individuals up to one month after administration of the Ronapreve (Casirivimab and Imdevimab) and/or Evusheld (Cilgavimab and Tixagevimab) antibody cocktails. All treated individuals displayed elevated antibody levels in their sera, which efficiently neutralized the Delta variant. Sera from Ronapreve recipients did not neutralize BA.1 and weakly inhibited BA.2. Neutralization of BA.1 and BA.2 was detected in 19 and 29 out of 29 Evusheld recipients, respectively. As compared to the Delta variant, neutralizing titers were more markedly decreased against BA.1 (344-fold) than BA.2 (9-fold). We further report 4 breakthrough Omicron infections among the 29 individuals, indicating that antibody treatment did not fully prevent infection. Collectively, BA.1 and BA.2 exhibit noticeable differences in their sensitivity to therapeutic mAbs. Anti-Omicron neutralizing activity of Ronapreve, and to a lesser extent that of Evusheld, is reduced in patients' sera.
Collapse
Affiliation(s)
- Timothée Bruel
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France. .,Vaccine Research Institute, Créteil, France.
| | - Jérôme Hadjadj
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP.CUP, Hôpital Cochin, Paris, France
| | - Piet Maes
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical and Epidemiological Virology, Leuven, Belgium
| | - Delphine Planas
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France.,Vaccine Research Institute, Créteil, France
| | - Aymeric Seve
- CHR d'Orléans, service de maladies infectieuses, Orléans, France
| | - Isabelle Staropoli
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France
| | | | - Françoise Porrot
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France
| | - William-Henry Bolland
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France.,Université de Paris, École doctorale BioSPC 562, Paris, France
| | - Yann Nguyen
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP.CUP, Hôpital Cochin, Paris, France
| | - Marion Casadevall
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP.CUP, Hôpital Cochin, Paris, France
| | - Caroline Charre
- Université de Paris, Faculté de Médecine, Paris, France.,INSERM U1016, CNRS UMR8104, Institut Cochin, Paris, France.,AP-HP, Laboratoire de Virologie, CHU Cochin, Paris, France
| | - Hélène Péré
- INSERM, Functional Genomics of Solid Tumors (FunGeST), Centre de Recherche des Cordeliers, Université de Paris and Sorbonne Université, Paris, France.,Laboratoire de Virologie, Service de Microbiologie, Hôpital Européen Georges Pompidou, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - David Veyer
- INSERM, Functional Genomics of Solid Tumors (FunGeST), Centre de Recherche des Cordeliers, Université de Paris and Sorbonne Université, Paris, France.,Laboratoire de Virologie, Service de Microbiologie, Hôpital Européen Georges Pompidou, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Matthieu Prot
- G5 Evolutionary Genomics of RNA Viruses, Institut Pasteur, Université de Paris, Paris, France
| | - Artem Baidaliuk
- G5 Evolutionary Genomics of RNA Viruses, Institut Pasteur, Université de Paris, Paris, France
| | - Lize Cuypers
- University Hospitals Leuven, Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, Leuven, Belgium
| | - Cyril Planchais
- Humoral Immunology Laboratory, Institut Pasteur, Université de Paris, INSERM U1222, Paris, France
| | - Hugo Mouquet
- Humoral Immunology Laboratory, Institut Pasteur, Université de Paris, INSERM U1222, Paris, France
| | - Guy Baele
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical and Epidemiological Virology, Leuven, Belgium
| | - Luc Mouthon
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP.CUP, Hôpital Cochin, Paris, France
| | | | - Etienne Simon-Loriere
- G5 Evolutionary Genomics of RNA Viruses, Institut Pasteur, Université de Paris, Paris, France
| | - Emmanuel André
- University Hospitals Leuven, Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, Leuven, Belgium.,KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, Leuven, Belgium
| | - Benjamin Terrier
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP.CUP, Hôpital Cochin, Paris, France
| | - Thierry Prazuck
- CHR d'Orléans, service de maladies infectieuses, Orléans, France
| | - Olivier Schwartz
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France. .,Vaccine Research Institute, Créteil, France.
| |
Collapse
|
30
|
Töllner M, Speer C, Benning L, Bartenschlager M, Nusshag C, Morath C, Zeier M, Süsal C, Schnitzler P, Schmitt W, Bergner R, Bartenschlager R, Lorenz HM, Schaier M. Impaired Neutralizing Antibody Activity against B.1.617.2 (Delta) after Anti-SARS-CoV-2 Vaccination in Patients Receiving Anti-CD20 Therapy. J Clin Med 2022; 11:jcm11061739. [PMID: 35330069 PMCID: PMC8952324 DOI: 10.3390/jcm11061739] [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: 02/08/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 12/19/2022] Open
Abstract
Background: To characterize humoral response after standard anti-SARS-CoV-2 vaccination in Rituximab-treated patients and to determine the optimal time point after last Rituximab treatment for appropriate immunization. Methods: Sixty-four patients who received Rituximab within the last seven years prior to the first anti-SARS-CoV-2 vaccination were recruited in a prospective observational study. Anti-S1 IgG, SARS-CoV-2 specific neutralization, and various SARS-CoV-2 target antibodies were determined. A live virus assay was used to assess neutralizing antibody activity against B.1.617.2 (delta). In Rituximab-treated patients, CD19+ peripheral B-cells were quantified using flow cytometry. Results: After second vaccination, all antibodies were significantly reduced compared to healthy controls. Neutralizing antibody activity against B.1.617.2 (delta) was detectable with a median (IQR) ID50 of 0 (0−1:20) compared to 1:320 (1:160−1:320) in healthy controls (for all p < 0.001). Longer time period since last Rituximab administration correlated with higher anti-SARS-CoV-2 antibody levels and a stronger neutralization of B.1.617.2 (delta). With one exception, only patients with a CD19+ cell proportion ≥ 1% had detectable neutralizing antibodies. Conclusion: Our data indicate that a reconstitution of the B-cell population to >1% seems crucial in developing neutralizing antibodies against SARS-CoV-2. We suggest that anti-SARS-CoV-2 vaccination should be administered at least 8−12 months after the last Rituximab treatment for sufficient humoral responses.
Collapse
Affiliation(s)
- Maximilian Töllner
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
- Correspondence:
| | - Claudius Speer
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
- Molecular Medicine Partnership Unit Heidelberg, European Molecular Biology Laboratory, 69120 Heidelberg, Germany
| | - Louise Benning
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| | - Marie Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, 69120 Heidelberg, Germany; (M.B.); (R.B.)
| | - Christian Nusshag
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| | - Christian Morath
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| | - Caner Süsal
- Transplant Immunology Research Center of Excellence, Koç University Hospital, Istanbul 34010, Turkey;
| | - Paul Schnitzler
- Department of Infectious Diseases, Virology, University of Heidelberg, 69120 Heidelberg, Germany;
| | | | - Raoul Bergner
- Clinical Center Ludwigshafen, Department of Internal Medicine A, 67036 Ludwigshafen, Germany;
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, 69120 Heidelberg, Germany; (M.B.); (R.B.)
- German Center for Infection Research (DZIF), Heidelberg Partner Site, 69120 Heidelberg, Germany
- Division Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Department of Medicine V, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Matthias Schaier
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| |
Collapse
|
31
|
Baron F, Canti L, Ariën KK, Kemlin D, Desombere I, Gerbaux M, Pannus P, Beguin Y, Marchant A, Humblet-Baron S. Insights From Early Clinical Trials Assessing Response to mRNA SARS-CoV-2 Vaccination in Immunocompromised Patients. Front Immunol 2022; 13:827242. [PMID: 35309332 PMCID: PMC8931657 DOI: 10.3389/fimmu.2022.827242] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/04/2022] [Indexed: 12/25/2022] Open
Abstract
It is critical to protect immunocompromised patients against COVID-19 with effective SARS-CoV-2 vaccination as they have an increased risk of developing severe disease. This is challenging, however, since effective mRNA vaccination requires the successful cooperation of several components of the innate and adaptive immune systems, both of which can be severely affected/deficient in immunocompromised people. In this article, we first review current knowledge on the immunobiology of SARS-COV-2 mRNA vaccination in animal models and in healthy humans. Next, we summarize data from early trials of SARS-COV-2 mRNA vaccination in patients with secondary or primary immunodeficiency. These early clinical trials identified common predictors of lower response to the vaccine such as anti-CD19, anti-CD20 or anti-CD38 therapies, low (naive) CD4+ T-cell counts, genetic or therapeutic Bruton tyrosine kinase deficiency, treatment with antimetabolites, CTLA4 agonists or JAK inhibitors, and vaccination with BNT162b2 versus mRNA1273 vaccine. Finally, we review the first data on third dose mRNA vaccine administration in immunocompromised patients and discuss recent strategies of temporarily holding/pausing immunosuppressive medication during vaccination.
Collapse
Affiliation(s)
- Frédéric Baron
- Laboratory of Hematology, GIGA-I3, University of Liege and Centre Hospitalier Universitaire (CHU) of Liège, Liege, Belgium
- Department of Medicine, Division of Hematology, Centre Hospitalier Universitaire (CHU) of Liège, Liège, Belgium
| | - Lorenzo Canti
- Laboratory of Hematology, GIGA-I3, University of Liege and Centre Hospitalier Universitaire (CHU) of Liège, Liege, Belgium
| | - Kevin K. Ariën
- Virology Unit, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Delphine Kemlin
- Department of Nephrology, Dialysis and Renal Transplantation, Hôpital Erasme, Université libre de Bruxelles, Brussels, Belgium
| | - Isabelle Desombere
- Scientific Directorate Infectious Diseases in Humans, Sciensano, Brussels, Belgium
| | - Margaux Gerbaux
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Université libre de Bruxelles (ULB), Gosselies, Belgium
- Department of Microbiology, Immunology and Transplantation, Laboratory of Adaptive Immunology, KU Leuven, Leuven, Belgium
| | - Pieter Pannus
- Scientific Directorate Infectious Diseases in Humans, Sciensano, Brussels, Belgium
| | - Yves Beguin
- Laboratory of Hematology, GIGA-I3, University of Liege and Centre Hospitalier Universitaire (CHU) of Liège, Liege, Belgium
- Department of Medicine, Division of Hematology, Centre Hospitalier Universitaire (CHU) of Liège, Liège, Belgium
| | - Arnaud Marchant
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Stéphanie Humblet-Baron
- Department of Microbiology, Immunology and Transplantation, Laboratory of Adaptive Immunology, KU Leuven, Leuven, Belgium
| |
Collapse
|
32
|
Sidler D, Born A, Schietzel S, Horn MP, Aeberli D, Amsler J, Möller B, Njue LM, Medri C, Angelillo-Scherrer A, Borradori L, Seyed Jafari SM, Radonjic-Hoesli S, Chan A, Hoepner R, Bacher U, Mani LY, Iype JM, Suter-Riniker F, Staehelin C, Nagler M, Hirzel C, Maurer B, Moor MB. Trajectories of humoral and cellular immunity and responses to a third dose of mRNA vaccines against SARS-CoV-2 in patients with a history of anti-CD20 therapy. RMD Open 2022; 8:e002166. [PMID: 35361691 PMCID: PMC8971359 DOI: 10.1136/rmdopen-2021-002166] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/09/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The majority of patients with B-cell-depleting therapies show compromised vaccination-induced immune responses. Herein, we report on the trajectories of anti-SARS-CoV-2 immune responses in patients of the RituxiVac study compared with healthy volunteers and investigate the immunogenicity of a third vaccination in previously humoral non-responding patients. METHODS We investigated the humoral and cell-mediated immune response after SARS-CoV-2 messanger RNA vaccination in patients with a history with anti-CD20 therapies. Coprimary outcomes were antispike and SARS-CoV-2-stimulated interferon-γ concentrations in vaccine responders 4.3 months (median; IQR: 3.6-4.8 months) after first evaluation, and humoral and cell-mediated immunity (CMI) after a third vaccine dose in previous humoral non-responders. Immunity decay rates were compared using analysis of covariance in linear regression. RESULTS 5.6 months (IQR: 5.1-6.7) after the second vaccination, we detected antispike IgG in 88% (29/33) and CMI in 44% (14/32) of patients with a humoral response after two-dose vaccination compared with 92% (24/26) healthy volunteers with antispike IgG and 69% (11/16) with CMI 6.8 months after the second vaccination (IQR: 6.0-7.1). Decay rates of antibody concentrations were comparable between patients and controls (p=0.70). In two-dose non-responders, a third SARS-CoV-2 vaccine elicited humoral responses in 19% (6/32) and CMI in 32% (10/31) participants. CONCLUSION This study reveals comparable immunity decay rates between patients with anti-CD20 treatments and healthy volunteers, but inefficient humoral or CMI after a third SARS-CoV-2 vaccine in most two-dose humoral non-responders calling for individually tailored vaccination strategies in this population.Trial registration numberNCT04877496; ClinicalTrials.gov number.
Collapse
Affiliation(s)
- Daniel Sidler
- Department of Nephrology and Hypertension, Inselspital University Hospital Bern, Bern, Switzerland
| | - Alexander Born
- Department of Nephrology and Hypertension, Inselspital University Hospital Bern, Bern, Switzerland
| | - Simeon Schietzel
- Department of Nephrology and Hypertension, Inselspital University Hospital Bern, Bern, Switzerland
| | - Michael P Horn
- Department of Clinical Chemistry, Inselspital Universitatsspital Bern, Bern, Switzerland
| | - Daniel Aeberli
- Department of Rheumatology and Immunology, Inselspital University Hospital Bern, Bern, Switzerland
| | - Jennifer Amsler
- Department of Rheumatology and Immunology, Inselspital University Hospital Bern, Bern, Switzerland
| | - Burkhard Möller
- Department of Rheumatology and Immunology, Inselspital University Hospital Bern, Bern, Switzerland
| | - Linet M Njue
- Department of Haematology and Central Haematology Laboratory, Inselspital University Hospital Bern, Bern, Switzerland
| | - Cesare Medri
- Department of Haematology and Central Haematology Laboratory, Inselspital University Hospital Bern, Bern, Switzerland
| | - Anne Angelillo-Scherrer
- Department of Haematology and Central Haematology Laboratory, Inselspital University Hospital Bern, Bern, Switzerland
| | - Luca Borradori
- Department of Dermatology, Inselspital University Hospital Bern, Bern, Switzerland
| | | | | | - Andrew Chan
- Department of Neurology, Inselspital University Hospital Bern, Bern, Switzerland
| | - Robert Hoepner
- Department of Neurology, Inselspital University Hospital Bern, Bern, Switzerland
| | - Ulrike Bacher
- Department of Haematology and Central Haematology Laboratory, Inselspital University Hospital Bern, Bern, Switzerland
| | - Laila-Yasmin Mani
- Department of Nephrology and Hypertension, Inselspital University Hospital Bern, Bern, Switzerland
| | - Joseena Mariam Iype
- Department of Clinical Chemistry, Inselspital Universitatsspital Bern, Bern, Switzerland
| | | | - Cornelia Staehelin
- Department of Infectious Diseases, Inselspital University Hospital Bern, Bern, Switzerland
| | - Michael Nagler
- Department of Clinical Chemistry, Inselspital Universitatsspital Bern, Bern, Switzerland
| | - Cedric Hirzel
- Department of Infectious Diseases, Inselspital University Hospital Bern, Bern, Switzerland
| | - Britta Maurer
- Department of Rheumatology and Immunology, Inselspital University Hospital Bern, Bern, Switzerland
| | - Matthias B Moor
- Department of Nephrology and Hypertension, Inselspital University Hospital Bern, Bern, Switzerland
| |
Collapse
|
33
|
Garcillán B, Salavert M, Regueiro JR, Díaz-Castroverde S. Response to Vaccines in Patients with Immune-Mediated Inflammatory Diseases: A Narrative Review. Vaccines (Basel) 2022; 10:297. [PMID: 35214755 PMCID: PMC8877652 DOI: 10.3390/vaccines10020297] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/26/2022] [Accepted: 02/13/2022] [Indexed: 12/28/2022] Open
Abstract
Patients with immune-mediated inflammatory diseases (IMIDs), such as rheumatoid arthritis and inflammatory bowel disease, are at increased risk of infection. International guidelines recommend vaccination to limit this risk of infection, although live attenuated vaccines are contraindicated once immunosuppressive therapy has begun. Biologic therapies used to treat IMIDs target the immune system to stop chronic pathogenic process but may also attenuate the protective immune response to vaccines. Here, we review the current knowledge regarding vaccine responses in IMID patients receiving treatment with biologic therapies, with a focus on the interleukin (IL)-12/23 inhibitors. B cell-depleting therapies, such as rituximab, strongly impair vaccines immunogenicity, and tumor necrosis factor (TNF) inhibitors and the cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) fusion protein abatacept are also associated with attenuated antibody responses, which are further diminished in patients taking concomitant immunosuppressants. On the other hand, integrin, IL-6, IL-12/23, IL-17, and B-cell activating factor (BAFF) inhibitors do not appear to affect the immune response to several vaccines evaluated. Importantly, treatment with biologic therapies in IMID patients is not associated with an increased risk of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or developing severe disease. However, the efficacy of SARS-CoV-2 vaccines on IMID patients may be reduced compared with healthy individuals. The impact of biologic therapies on the response to SARS-CoV-2 vaccines seems to replicate what has been described for other vaccines. SARS-CoV-2 vaccination appears to be safe and well tolerated in IMID patients. Attenuated but, in general, still protective responses to SARS-CoV-2 vaccination in the context of certain therapies warrant current recommendations for a third primary dose in IMID patients treated with immunosuppressive drugs.
Collapse
Affiliation(s)
| | - Miguel Salavert
- Infectious Disease Unit, Department of Clinical Medicine, La Fe Health Research Institute, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain;
| | - José R. Regueiro
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, 12 de Octubre Health Research Institute (imas12), 28040 Madrid, Spain;
| | | |
Collapse
|