1
|
Wroński J, Massalska M, Jaszczyk B, Felis-Giemza A, Kornatka A, Plebańczyk M, Burakowski T, Kwiatkowska B, Kuca-Warnawin E, Ciechomska M. Beyond interferon gamma - decreased cellular response to COVID-19 vaccination booster in patients with autoimmune inflammatory rheumatic diseases. Front Immunol 2025; 16:1568439. [PMID: 40226625 PMCID: PMC11986637 DOI: 10.3389/fimmu.2025.1568439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
The global COVID-19 pandemic has led to significant advancements in vaccine research, particularly regarding patients with autoimmune inflammatory rheumatic diseases (AIIRD). However, most studies have assessed the post-vaccination cellular response only by measuring the production of interferon-gamma. This study aimed to explore the post-vaccination cellular immune response in patients with AIIRD, with a focus on the effects of immunomodulatory drugs on different proteins involved in the cellular response and cytotoxicity. We analyzed blood samples from 54 patients - 16 healthy controls (HC) and 38 AIIRD patients - at three time points: before (T0), 4 weeks after (T1), and more than 6 months after (T2) a COVID-19 booster vaccination. Gene expression and concentration levels of 13 proteins involved in cellular immunity were assessed. Our study showed significantly reduced production of TNF at T0, IL-2 at T0 and T2, and perforin at T2 in AIIRD patients compared to HC. In AIIRD patients the expression of genes involved in cytotoxicity, including NRF2, TRAIL, cathepsin B, and cathepsin H was impaired. Both protein concentrations and gene expression were particularly altered in those treated with glucocorticoids, methotrexate, and biologic/targeted synthetic disease-modifying antirheumatic drugs (b/tsDMARDs). Among b/tsDMARDs only IL-17 inhibitors did not affect the cellular response. These findings suggest that COVID-19 vaccination elicits a weakened cellular response in patients with AIIRD, particularly those on immunosuppressive therapies, potentially compromising vaccine efficacy. Further studies are required to determine the clinical impact of these findings on long-term vaccine effectiveness in this population.
Collapse
Affiliation(s)
- Jakub Wroński
- Department of Rheumatology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Magdalena Massalska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Bożena Jaszczyk
- Department of Outpatient Clinics, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Anna Felis-Giemza
- Biologic Therapy Center, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Anna Kornatka
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Magdalena Plebańczyk
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Tomasz Burakowski
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Brygida Kwiatkowska
- Department of Early Arthritis, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Ewa Kuca-Warnawin
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Marzena Ciechomska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| |
Collapse
|
2
|
Keijser JBD, Stalman EW, Wieske L, Steenhuis M, van Dam KPJ, Kummer LYL, van Kempen ZLE, Killestein J, Volkers AG, Tas SW, Boekel L, Wolbink GJ, Blanco LF, Verstegen NJM, Keijzer S, van Mierlo G, Cristianawati O, Boogaard AJ, van der Straten K, van Rijswijk J, van Gils MJ, Ten Brinke A, van Ham SM, Kuijpers TW, Eftimov F, Rispens T. Evolution of SARS-CoV-2 antibody repertoire after successive mRNA vaccinations under immunosuppressive treatment. EBioMedicine 2025; 113:105620. [PMID: 40010153 PMCID: PMC11905820 DOI: 10.1016/j.ebiom.2025.105620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Repeated antigen exposure can result in a shifting antibody repertoire. The mechanisms by which this occurs and consequences for cross-variant protection against evolving pathogens remain incompletely understood, particularly in the context of immunosuppressive treatments used in patients with immune-mediated inflammatory diseases (IMID). METHODS To investigate this, we characterised longitudinal changes in the anti-SARS-CoV-2 antibody repertoire over the course of three SARS-CoV-2 mRNA vaccinations in patients with IMIDs treated with methotrexate (MTX) and/or tumour necrosis factor-inhibitors (TNFi), anti-CD20 monoclonal antibodies, no systemic therapy, and healthy controls (total N = 878). We determined serum antibody titres against the receptor-binding domain (RBD) of Wuhan-Hu-1 (WH1) and Omicron BA.1 spike proteins, and assessed ratios thereof between groups as a proxy for cross-reactivity. FINDINGS We observe emerging anti-BA.1 RBD reactivity over time, notably following a third vaccination. This may be partly explained by affinity maturation, as evaluated by inhibition of ACE2-RBD interactions. Similar trends were seen in patients treated with MTX and/or TNFi, but not in patients on anti-CD20 therapy. SARS-CoV-2 infection prior to vaccination accelerated these effects initially while leading to comparable results after three vaccinations. INTERPRETATION MTX and TNFi do not qualitatively alter the evolution of the antibody repertoire in response to repeated antigen exposure, whereas anti-CD20 does. These insights may help to optimise vaccination strategies for patients with immune-mediated inflammatory diseases. FUNDING This study was supported by ZonMw (The Netherlands Organization for Health Research and Development) and SGF (Collaborating Health Funds).
Collapse
Affiliation(s)
- Jim B D Keijser
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Eileen W Stalman
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Luuk Wieske
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Clinical Neurophysiology, St Antonius Hospital, Nieuwegein, the Netherlands
| | - Maurice Steenhuis
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Koos P J van Dam
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Laura Y L Kummer
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands; Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Joep Killestein
- Department of Neurology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Adriaan G Volkers
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology and Clinical Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Laura Boekel
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology, Reade, Amsterdam, the Netherlands
| | - Gerrit J Wolbink
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology, Reade, Amsterdam, the Netherlands
| | - Laura Fernandez Blanco
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Niels J M Verstegen
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Sofie Keijzer
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Gerard van Mierlo
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Olvi Cristianawati
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Arend J Boogaard
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Karlijn van der Straten
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands; Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jacqueline van Rijswijk
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands; Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Marit J van Gils
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands; Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Anja Ten Brinke
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands; Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Taco W Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Filip Eftimov
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands; Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Vlădulescu-Trandafir AI, Bojincă VC, Munteanu C, Anghelescu A, Popescu C, Stoica SI, Aurelian S, Bălănescu A, Băetu C, Ciobanu V, Onose G. Rheumatoid Arthritis and COVID-19 at the Intersection of Immunology and Infectious Diseases: A Related PRISMA Systematic Literature Review. Int J Mol Sci 2024; 25:11149. [PMID: 39456932 PMCID: PMC11508285 DOI: 10.3390/ijms252011149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Rheumatoid arthritis (RA) patients face different health challenges when infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) than the general population, due to both their immunocompromised state and the immunosuppressive therapies they receive. This systematic literature review, which follows the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) paradigm, explores the interactions between RA and SARS-CoV-2 infection, focusing on immunologic issues, disease management, vaccination, and adverse outcomes. In order to obtain the most relevant information, we systematically reviewed the specific literature from 1 January 2021 to 31 December 2023, based on the PRISMA method, by which we eventually selected 35 eligible articles, to which we added other ISI-indexed studies to enrich our results further. Consequently, we performed a funnel analysis to evaluate the potential for publication bias. Firstly, the data collected revealed the impact of the pandemic on RA diagnoses and the fear of face-to-face medical consultations that delayed adequate treatment. Secondly, cardiovascular and metabolic comorbidities increase the risk of prolonged COVID-19 symptoms, hospitalization, and severe COVID-19 outcomes for RA patients. With respect to immunosuppressive treatment used to control RA, it was observed that glucocorticoids (especially high-dose usage) and Rituximab (RTX) predispose the patients to poor SARS-CoV-2 outcomes, as opposed to Baricitinib and interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) inhibitors. COVID-19 vaccination has proven effective and generally safe for RA patients in some studies, although therapies with Methotrexate (MTX), Abatacept (ABA), and RTX have been associated with impaired vaccine immune response. This systematic literature review brings updated and thorough information with respect to the immunological, clinical, and management of a complex immune-mediated inflammatory disease (IMID) like RA in the setting of COVID-19 and underlines the challenges faced by this group of patients. The lessons learned can be extended beyond the pandemic in shaping a more informed and compassionate healthcare system and offering long-term medical care for patients with RA.
Collapse
Affiliation(s)
- Andreea-Iulia Vlădulescu-Trandafir
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (A.-I.V.-T.); (V.-C.B.); (S.A.); (A.B.); (C.B.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.A.); (S.-I.S.)
| | - Violeta-Claudia Bojincă
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (A.-I.V.-T.); (V.-C.B.); (S.A.); (A.B.); (C.B.); (G.O.)
- Internal Medicine and Rheumatology Departments, “Sfânta Maria” Hospital, 011172 Bucharest, Romania
| | - Constantin Munteanu
- Neuromuscular Rehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.A.); (S.-I.S.)
- Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa” Iasi, 700454 Iasi, Romania
| | - Aurelian Anghelescu
- Neuromuscular Rehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.A.); (S.-I.S.)
- Faculty of Midwifery and Nursing, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Cristina Popescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (A.-I.V.-T.); (V.-C.B.); (S.A.); (A.B.); (C.B.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.A.); (S.-I.S.)
| | - Simona-Isabelle Stoica
- Neuromuscular Rehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.A.); (S.-I.S.)
- Faculty of Midwifery and Nursing, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Sorina Aurelian
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (A.-I.V.-T.); (V.-C.B.); (S.A.); (A.B.); (C.B.); (G.O.)
- Gerontology and Geriatrics Clinic Division, St. Luca Hospital for Chronic Illnesses, 041915 Bucharest, Romania
| | - Andra Bălănescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (A.-I.V.-T.); (V.-C.B.); (S.A.); (A.B.); (C.B.); (G.O.)
- Internal Medicine and Rheumatology Departments, “Sfânta Maria” Hospital, 011172 Bucharest, Romania
| | - Cristina Băetu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (A.-I.V.-T.); (V.-C.B.); (S.A.); (A.B.); (C.B.); (G.O.)
- Neurology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Vlad Ciobanu
- Computer Science Department, Politehnica University of Bucharest, 060042 Bucharest, Romania;
| | - Gelu Onose
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (A.-I.V.-T.); (V.-C.B.); (S.A.); (A.B.); (C.B.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.A.); (S.-I.S.)
| |
Collapse
|
4
|
Alonzi T, Aiello A, Sali M, Delogu G, Villella VR, Raia V, Nicastri E, Piacentini M, Goletti D. Multiple antimicrobial and immune-modulating activities of cysteamine in infectious diseases. Biomed Pharmacother 2024; 178:117153. [PMID: 39024833 DOI: 10.1016/j.biopha.2024.117153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024] Open
Abstract
Infectious diseases are a major threat to global health and cause millions of deaths every year, particularly in developing countries. The emergence of multidrug resistance challenges current antimicrobial treatments, inducing uncertainty in therapeutic protocols. New compounds are therefore necessary. A drug repurposing approach could play a critical role in developing new treatments used either alone or in combination with standard therapy regimens. Herein, we focused on cysteamine, an aminothiol endogenously synthesized by human cells during the degradation of coenzyme-A, which is a drug approved for the treatment of nephropathic cystinosis. Cysteamine influences many biological processes due to the presence of the highly reactive thiol group. This review provides an overview of cysteamine-mediated effects on different viruses, bacteria and parasites, with a particular focus on infections caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), Mycobacterium tuberculosis, non-tuberculous mycobacteria (NTM), and Pseudomonas aeruginosa. Evidences for a potential use of cysteamine as a direct antimicrobial agent and/or a host-directed therapy, either alone or in combination with other antimicrobial drugs, are described.
Collapse
Affiliation(s)
- Tonino Alonzi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Alessandra Aiello
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Michela Sali
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, India; Dipartimento di Scienze di Laboratorio ed Ematologiche, Fondazione Policlinico Universitario A. Gemelli, IRCCS; Rome, Italy
| | - Giovanni Delogu
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, India; Mater Olbia Hospital, Olbia, Italy
| | - Valeria Rachela Villella
- CEINGE, Advanced Biotechnologies Franco Salvatore, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Valeria Raia
- Pediatric Unit, Department of Translational Medical Sciences, Regional Cystic Fibrosis Center, Federico II University Naples, Naples, Italy
| | - Emanuele Nicastri
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy; Cell Biology and Electron Microscopy Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy.
| |
Collapse
|
5
|
Sato H, Nagatani K, Minota S. Diminished antibody response to SARS-CoV-2 in rheumatoid arthritis compared to metabolic disorders following the primary series of vaccinations and its recovery with a booster: A single-center prospective observational study. Int J Rheum Dis 2024; 27:e15203. [PMID: 38924257 DOI: 10.1111/1756-185x.15203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 06/28/2024]
Abstract
AIM To compare antibody responses after vaccinations between patients with rheumatoid arthritis (RA) and patients with metabolic disorders (MD). The study places special emphasis on understanding how common diseases affect antibody responses in individuals with RA within real-world settings. METHODS The participants were 117 patients with RA (66 with RA only and 51 with RA and MD) and 37 patients with MD who received both the primary series of vaccinations and a booster. Antibody titers were compared after the primary series of vaccinations and a booster, and factors influencing the antibody response were assessed. RESULTS Following the primary series of vaccinations, a significant reduction in antibody titers was observed in patients with longer days between vaccination and antibody measurement, the use of IL-6 inhibitors, selective T cell co-stimulation modulators, and methotrexate. Comorbid MD did not exhibit significant influences on antibody response in RA. Notably, the presence of RA itself was not significant in multivariate linear regression analysis. After the administration of the booster, however, day between vaccination and antibody measurement, the use of IL-6 inhibitor, and methotrexate no longer remained significant. Only the use of selective T cell co-stimulation modulators retained its significance. CONCLUSIONS MD did not exhibit a significant impact on antibody responses in RA patients. The reduced antibody response following the primary series in RA patients appeared to be attributed more to specific RA medications rather than to the disease itself. Booster vaccines are vital in restoring the antibody response in RA.
Collapse
Affiliation(s)
| | - Katsuya Nagatani
- Division of Rheumatology and Clinical Immunology, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Seiji Minota
- Nasu-Kogen Clinic, Nasu-Shiobara, Tochigi, Japan
| |
Collapse
|
6
|
Kong X, Wang W. Coronavirus disease 2019 vaccination in patients with rheumatic diseases: A bibliometric‐based analysis of trends. RHEUMATOLOGY & AUTOIMMUNITY 2024; 4:81-89. [DOI: 10.1002/rai2.12132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 04/10/2024] [Indexed: 01/03/2025]
Abstract
AbstractBackgroundCoronavirus disease 2019 (COVID‐19) vaccination in patients with rheumatic disease is of practical clinical importance. This study aimed to perform a comprehensive bibliometric analysis of COVID‐19 vaccination in patients with rheumatic disease and indicate possible directions for future studies.MethodsResearch articles and reviews related to COVID‐19 vaccinations in patients with rheumatic diseases were retrieved from Science Citation Index Expanded of the Web of Science Core Collection. The CiteSpace and VOSviewer software were used to depict network mapping showing the collaborations among countries, institutions, and authors. Current hotspots and future directions were derived by analyzing highly cited/co‐cited documents and keywords.ResultsIn total, 615 documents published in 194 academic journals, including 505 articles and 110 reviews contributed by 5068 authors from 1693 institutions in 79 countries/regions, were finally included and analyzed. The United States and the University of London were the most productive country and cooperative institution, respectively. Author analysis showed that cooperation between different authors was largely confined to only several groups. Vaccines and Journal of the American Academy of Dermatology were the journals with the most publications and citations per document, respectively. Cluster analysis showed that the keywords can be categorized into groups like kidney disease, antibody response, COVID‐19 vaccine, vaccination, validation, safety, and giant cell arteritis.ConclusionsThe efficacy and safety of COVID‐19 vaccination in patients with rheumatic disease are being continuously investigated. Future studies on COVID‐19 vaccination in patients with rheumatic diseases could focus on antibody response, validation, and vaccine safety.
Collapse
Affiliation(s)
- Xiufang Kong
- Department of Rheumatology, Zhongshan Hospital Fudan University Shanghai China
| | - Wei Wang
- Department of Nephrology Shanghai Tenth People's Hospital Shanghai China
| |
Collapse
|
7
|
Sagawa F, Yamada H, Ayano M, Kimoto Y, Mitoma H, Ono N, Arinobu Y, Kondo M, Nakashima Y, Akashi K, Horiuchi T, Niiro H. Determination of the factors associated with antigen-specific CD4+ T-cell responses to BNT162b2 in patients with rheumatoid arthritis. RMD Open 2024; 10:e003693. [PMID: 38216287 PMCID: PMC10806600 DOI: 10.1136/rmdopen-2023-003693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/19/2023] [Indexed: 01/14/2024] Open
Abstract
OBJECTIVES Understanding interpatient variation in CD4+T-cell responses is the bases for understanding the pathogenesis and management of rheumatoid arthritis (RA). We examined immune responses to SARS-CoV-2 vaccine in a cohort of patients with RA and determined factors associated with the responses. METHODS Four hundred and thirty-one patients with RA having received two doses of BNT162b2, a messenger RNA-based vaccine for SARS-CoV-2, were included. Vaccine antigen-specific IgG was detected by ELISA, and antigen-specific CD4+T cells were detected by CD154 expression in response to antigenic stimulation. Expression of cytokines was concomitantly detected by intracellular staining. Associations among background variables, antigen-specific antibody production and the CD4+T-cell responses were analysed. Unsupervised hierarchical clustering was performed based on the profiles of antigen-specific cytokine production by CD4+T cells to stratify patients with RA. RESULTS Multivariate analysis indicated that ageing negatively affects CD4+T-cell response as well as antibody production. No association was detected between the presence or the levels of rheumatoid factor/anti-cyclic citrullinated peptide antibody and anti-vaccine immune responses. Methotrexate and prednisolone reduced B cell but not T-cell responses. Conventional immunophenotyping by the expression of chemokine receptors was not associated with the actual CD4+T-cell response, except for T helper cells (Th1). Functional immunophenotyping based on the profiles of antigen-specific cytokine production of CD4+T cells stratified patients with RA into three clusters, among which Th1-dominant type less frequently underwent joint surgery. CONCLUSIONS Clinical and immunological variables that are associated with antigen-specific CD4 T-cell responses in patients with RA were determined by analysing immune responses against SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- Fumiaki Sagawa
- Department of Medicine and Biosystemic Science, Kyushu University, Fukuoka, Japan
| | - Hisakata Yamada
- Department of Clinical Immunology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masahiro Ayano
- Department of Medicine and Biosystemic Science, Kyushu University, Fukuoka, Japan
| | - Yasutaka Kimoto
- Department of Internal Medicine, Kyushu University Beppu Hospital, Beppu, Japan
| | - Hiroki Mitoma
- Department of Medicine and Biosystemic Science, Kyushu University, Fukuoka, Japan
| | - Nobuyuki Ono
- Department of Medicine and Biosystemic Science, Kyushu University, Fukuoka, Japan
| | - Yojiro Arinobu
- Department of Medicine and Biosystemic Science, Kyushu University, Fukuoka, Japan
| | - Masakazu Kondo
- Kondo Clinic for Rheumatology and Orthopaedics, Fukuoka, Japan
| | | | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University, Fukuoka, Japan
| | | | - Hiroaki Niiro
- Department of Medical Education, Kyushu University, Fukuoka, Japan
| |
Collapse
|
8
|
van Sleen Y, van der Geest KSM, Buisman AM, Sandovici M, van Baarle D, Brouwer E. Humoral SARS-CoV-2 Vaccine Responses in Patients With Giant Cell Arteritis and Polymyalgia Rheumatica: Decay After Primary Vaccination and Effects of the Booster. Arthritis Care Res (Hoboken) 2024; 76:105-110. [PMID: 37332051 DOI: 10.1002/acr.25173] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/11/2023] [Accepted: 06/13/2023] [Indexed: 06/20/2023]
Abstract
OBJECTIVE Vaccination remains essential in preventing morbidity of SARS-CoV-2 infections. We previously showed that >10 mg/day of prednisolone and methotrexate was associated with reduced antibody concentrations after primary vaccination in patients with giant cell arteritis (GCA) and polymyalgia rheumatica (PMR). This follow-up study was undertaken to measure the decay of antibody concentrations and the immunogenicity of SARS-CoV-2 booster vaccination. METHODS Patients with GCA/PMR included in the primary vaccination (BNT162b2 [Pfizer-BioNTech] or ChAdOx1 [Oxford/AstraZeneca]) study were asked again to donate blood samples 6 months after primary vaccination (n = 24) and 1 month after booster vaccination (n = 46, BNT162b2 or mRNA1273). Data were compared to those of age-, sex-, and vaccine-matched controls (n = 58 and n = 42, respectively). Multiple linear regression was performed with post-booster antibody concentrations as dependent variable and post-primary vaccination antibodies, prednisolone >10mg/day, and methotrexate use as predicting variables. RESULTS Antibody concentrations decreased faster over time in GCA/PMR patients than in controls, which was associated with prednisolone treatment during primary vaccination. Post-booster antibody concentrations were comparable between patients and controls. Antibody concentrations post primary vaccination, but not treatment during booster vaccination, were predictive for antibody concentrations post booster vaccination. CONCLUSION These results indicate that the decay of humoral immunity after primary vaccination is associated with prednisolone treatment, whereas the subsequent increase after booster vaccination, was not. Patients with low antibody concentrations following primary vaccination remained at an immunogenic disadvantage after a single booster vaccination. This longitudinal study in GCA/PMR patients stresses the importance of repeated booster vaccination for patients with poor responses to primary vaccination.
Collapse
Affiliation(s)
| | | | - Anne-Marie Buisman
- National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Maria Sandovici
- University Medical Center Groningen, Groningen, The Netherlands
| | | | | |
Collapse
|
9
|
Isnardi CA, Landi M, Cruces L, Maid P, Calle Montoro C, Alfaro MA, Roldán BM, Gómez Vara AB, Giorgis P, Ezquer RA, Crespo Rocha MG, Reyes Gómez CR, Correa MÁ, Cerda OL, Rosemffet MG, Carrizo Abarza V, Catalan Pellet S, Perandones M, Reimundes C, Longueira Y, Turk G, Quiroga MF, Laufer N, De La Vega MC, Citera G, Pons-Estel GJ, Schneeberger EE. Humoral and T Cell Response to SARS-CoV-2 Vaccination in Patients With Rheumatoid Arthritis. Arthritis Care Res (Hoboken) 2024; 76:120-130. [PMID: 37605835 DOI: 10.1002/acr.25221] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 08/02/2023] [Accepted: 08/18/2023] [Indexed: 08/23/2023]
Abstract
OBJECTIVE The objective of this study was to assess the SARS-CoV-2-specific humoral and T cell response after a two-dose regimen of SARS-CoV-2 vaccine in patients with rheumatoid arthritis (RA). METHODS In this observational study, patients with RA who are ≥18 years of age and vaccinated for SARS-CoV-2 according to the Argentine National Health Ministry's vaccination strategy were included. Anti-SARS-CoV-2 immunoglobulin G (IgG) antibodies (ELISA-COVIDAR test), neutralizing activity (cytotoxicity in VERO cells), and specific T cell response (IFN-γ ELISpot Assay) were assessed after the first and second dose. RESULTS A total of 120 patients with RA were included. Mostly, homologous regimens were used, including Gam-COVID-Vac (27.5%), ChAdOx1 (24.2%), and BBIBP-CorV (22.5%). The most frequent combination was Gam-COVID-Vac/mRNA-1273 (21.7%). After the second dose, 81.7% presented with anti-SARS-CoV-2 antibodies, 70.0% presented with neutralizing activity, and 65.3% presented with specific T cell response. The use of BBIBP-CorV and treatment with abatacept (ABA) and rituximab (RTX) were associated with undetectable antibodies and no neutralizing activity after two doses. BBIBP-CorV was also associated with the absence of T cell response. The total incidence of adverse events was 357.1 events per 1,000 doses, significantly lower with BBIBP-CorV (166.7 events per 1,000 doses, P < 0.02). CONCLUSION In this RA cohort vaccinated with homologous and heterologous regimens against COVID-19, 2 out of 10 patients did not develop anti-SARS-CoV-2 IgG, 70% presented with neutralizing activity, and 65% presented with specific T cell response. The use of BBIBP-CorV was associated with deficient humoral and cellular response, whereas treatment with ABA and RTX resulted in an impaired anti-SARS-CoV-2 IgG formation and neutralizing activity.
Collapse
Affiliation(s)
- Carolina A Isnardi
- Research Unit of the Argentine Society of Rheumatology, Ciudad Autónoma de Buenos Aires, Argentina
| | - Margarita Landi
- Research Unit of the Argentine Society of Rheumatology, Ciudad Autónoma de Buenos Aires, Argentina
| | - Leonel Cruces
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Reetrovirus y SIDA (INBIRS), Universisdad de Buenos Aires, facultad de Medicina, Ciudad Autónoma de Buenos Aires, Argentina
| | - Pablo Maid
- Hospital Universitario Austral, Buenos Aires, Argentina
| | | | - María A Alfaro
- Instituto de Rehabilitación Psicofísica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Brian M Roldán
- Instituto de Rehabilitación Psicofísica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Andrea B Gómez Vara
- Instituto de Rehabilitación Psicofísica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Pamela Giorgis
- Instituto de Rehabilitación Psicofísica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Roberto A Ezquer
- Instituto de Rehabilitación Psicofísica, Ciudad Autónoma de Buenos Aires, Argentina
| | - María G Crespo Rocha
- Instituto de Rehabilitación Psicofísica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Camila R Reyes Gómez
- Instituto de Rehabilitación Psicofísica, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Á Correa
- Instituto de Rehabilitación Psicofísica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Osvaldo L Cerda
- Instituto de Rehabilitación Psicofísica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Marcos G Rosemffet
- Instituto de Rehabilitación Psicofísica, Ciudad Autónoma de Buenos Aires, Argentina
| | | | | | - Miguel Perandones
- Instituto de Rehabilitación Psicofísica, Ciudad Autónoma de Buenos Aires, Argentina
| | | | - Yesica Longueira
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Reetrovirus y SIDA (INBIRS), Universisdad de Buenos Aires, facultad de Medicina, Ciudad Autónoma de Buenos Aires, Argentina
| | - Gabriela Turk
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Reetrovirus y SIDA (INBIRS), Universisdad de Buenos Aires, facultad de Medicina, Ciudad Autónoma de Buenos Aires, Argentina
| | - María F Quiroga
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Reetrovirus y SIDA (INBIRS), Universisdad de Buenos Aires, facultad de Medicina, Ciudad Autónoma de Buenos Aires, Argentina
| | - Natalia Laufer
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Reetrovirus y SIDA (INBIRS), Universisdad de Buenos Aires, facultad de Medicina, Ciudad Autónoma de Buenos Aires, Argentina
| | - María C De La Vega
- Argentine Society of Rheumatology, Ciudad Autónoma de Buenos Aires, Argentina
| | - Gustavo Citera
- Instituto de Rehabilitación Psicofísica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Guillermo J Pons-Estel
- Research Unit of the Argentine Society of Rheumatology, Ciudad Autónoma de Buenos Aires, Argentina
| | | |
Collapse
|
10
|
Benoit JM, Breznik JA, Ang JC, Bhakta H, Huynh A, Cowbrough B, Baker B, Heessels L, Lodhi S, Yan E, Ewusie J, Nazy I, Bramson J, Miller MS, Bernatsky S, Larché MJ, Bowdish DME. Immunomodulatory drugs have divergent effects on humoral and cellular immune responses to SARS-CoV-2 vaccination in people living with rheumatoid arthritis. Sci Rep 2023; 13:22846. [PMID: 38129594 PMCID: PMC10739702 DOI: 10.1038/s41598-023-50263-5] [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: 07/12/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023] Open
Abstract
Understanding the efficacy of SARS-CoV-2 vaccination in people on immunosuppressive drugs, including those with rheumatoid arthritis (RA), is critical for their protection. Vaccine induced protection requires antibodies, CD4+ T cells, and CD8+ T cells, but it is unclear if these are equally affected by immunomodulatory drugs. Here, we determined how humoral and cellular SARS-CoV-2 vaccination responses differed between people with RA and controls, and which drug classes impacted these responses. Blood was collected from participants with RA on immunomodulatory drugs and controls after their second, third, and fourth SARS-CoV-2 vaccinations. Receptor binding domain (RBD)-specific antibodies were quantified by ELISA. Spike-specific memory T cells were quantitated using flow cytometry. Linear mixed models assessed the impact of age, sex, and immunomodulatory drug classes on SARS-CoV-2 vaccination responses. Compared to non-RA controls (n = 35), participants with RA on immunomodulatory drugs (n = 62) had lower anti-RBD IgG and spike-specific CD4+ T cell levels, but no deficits in spike-specific CD8+ T cells, following SARS-CoV-2 vaccination. Use of costimulation inhibitors was associated with lower humoral responses. JAK inhibitors were associated with fewer spike-specific CD4+ T cells. Participants with RA on immunomodulatory drugs mounted weaker responses to SARS-CoV-2 vaccination, with different drug classes impacting the cellular and humoral compartments.
Collapse
Affiliation(s)
- Jenna M Benoit
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
- Firestone Institute of Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Jessica A Breznik
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
- McMaster Institute for Research on Aging, McMaster University, Hamilton, ON, Canada
| | - Jann C Ang
- Department of Biochemistry, McMaster University, Hamilton, ON, Canada
| | - Hina Bhakta
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Angela Huynh
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Braeden Cowbrough
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
| | - Barbara Baker
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Lauren Heessels
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Sumiya Lodhi
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Elizabeth Yan
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Joycelyne Ewusie
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, ON, Canada
| | - Ishac Nazy
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jonathan Bramson
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
| | - Matthew S Miller
- McMaster Immunology Research Centre, Hamilton, ON, Canada
- Department of Biochemistry, McMaster University, Hamilton, ON, Canada
| | - Sasha Bernatsky
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Maggie J Larché
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
- McMaster Immunology Research Centre, Hamilton, ON, Canada.
| | - Dawn M E Bowdish
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
- McMaster Immunology Research Centre, Hamilton, ON, Canada.
- Firestone Institute of Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada.
| |
Collapse
|
11
|
Lee JM, Figueroa A, Sachithanandham J, Li M, Connolly CM, Shapiro JR, Chen Y, Jones M, Dhara VG, Towns M, Lee JS, Peralta SR, Milstone AM, Betenbaugh M, Debes AK, Blankson J, Sitaras I, Yoon S, Thompson EA, Bingham CO, Klein SL, Pekosz A, Bailey JR. Three doses of COVID-19 mRNA vaccine induce class-switched antibody responses in inflammatory arthritis patients on immunomodulatory therapies. Front Immunol 2023; 14:1266370. [PMID: 38022602 PMCID: PMC10646683 DOI: 10.3389/fimmu.2023.1266370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
Patients with inflammatory arthritis (IA) are at increased risk of severe COVID-19 due to medication-induced immunosuppression that impairs host defenses. The aim of this study was to assess antibody and B cell responses to COVID-19 mRNA vaccination in IA patients receiving immunomodulatory therapies. Adults with IA were enrolled through the Johns Hopkins Arthritis Center and compared with healthy controls (HC). Paired plasma and peripheral blood mononuclear cell (PBMC) samples were collected prior to and 30 days or 6 months following the first two doses of mRNA vaccines (D2; HC=77 and IA=31 patients), or 30 days following a third dose of mRNA vaccines (D3; HC=11 and IA=96 patients). Neutralizing antibody titers, total binding antibody titers, and B cell responses to vaccine and Omicron variants were analyzed. Anti-Spike (S) IgG and S-specific B cells developed appropriately in most IA patients following D3, with reduced responses to Omicron variants, and negligible effects of medication type or drug withholding. Neutralizing antibody responses were lower compared to healthy controls after both D2 and D3, with a small number of individuals demonstrating persistently undetectable neutralizing antibody levels. Most IA patients respond as well to mRNA COVID-19 vaccines as immunocompetent individuals by the third dose, with no evidence of improved responses following medication withholding. These data suggest that IA-associated immune impairment may not hinder immunity to COVID-19 mRNA vaccines in most individuals.
Collapse
Affiliation(s)
- Jenny M. Lee
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Alexis Figueroa
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jaiprasath Sachithanandham
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Maggie Li
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Caoilfhionn M. Connolly
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Janna R. Shapiro
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Yiqun Chen
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Michelle Jones
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Venkata Gayatri Dhara
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Marilyn Towns
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - John S. Lee
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Stephanie R. Peralta
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Aaron M. Milstone
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Bloomberg School of Public Health, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Amanda K. Debes
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Joel Blankson
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ioannis Sitaras
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Steve Yoon
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Elizabeth A. Thompson
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Clifton O. Bingham
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Justin R. Bailey
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
12
|
Limoges MA, Lortie A, Demontier É, Quenum AJI, Lessard F, Drouin Z, Carrier N, Nguimbus LM, Beaulieu MC, Boire G, Piché A, Allard-Chamard H, Ramanathan S, Roux S. SARS-CoV-2 mRNA vaccine-induced immune responses in rheumatoid arthritis. J Leukoc Biol 2023; 114:358-367. [PMID: 37478373 PMCID: PMC10533224 DOI: 10.1093/jleuko/qiad086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 07/15/2023] [Accepted: 07/19/2023] [Indexed: 07/23/2023] Open
Abstract
Our objective was to characterize T and B cell responses to vaccination with SARS-CoV-2 antigens in immunocompromised rheumatoid arthritis (RA) patients. In 22 RA patients, clinical and biological variables were analyzed before and 4 weeks after each of 3 messenger RNA (mRNA) vaccine doses and compared with unmatched healthy individuals. Sequentially sampled peripheral blood mononuclear cells and sera were collected to determine immune profiles and to analyze the T cell response to a spike peptide pool and B cell specificity to the receptor-binding domain (RBD). Anti-spike antibodies were detectable in 6 of 22 RA patients after 1 dose of vaccine with increasing titers after each booster dose, although the overall response was lower compared with that in healthy control individuals. Responding patients after the first dose were more likely to have RA antibodies and a higher baseline proportion of circulating follicular B cells. In RA patients, the mRNA vaccine elicited a robust CD4+ T response to a spike peptide pool following the first and second doses. Consistent with the serologies, RBD-specific B cells exhibited a modest increase after the first dose and the second dose resulted in marked increases only in a fraction of the RA patients to both ancestral and omicron RBD. Our results highlight the importance of multidose COVID-19 vaccination in RA patients to develop a protective humoral response. However, these patients rapidly develop specific T CD4+ responses, despite delayed B cell responses.
Collapse
Affiliation(s)
- Marc-André Limoges
- Department of Immunology and Cell Biology, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| | - Audrey Lortie
- Division of Rheumatology, Department of Medicine, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| | - Élodie Demontier
- Division of Rheumatology, Department of Medicine, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| | - Akouavi Julite Irmine Quenum
- Department of Immunology and Cell Biology, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| | - Félix Lessard
- Division of Rheumatology, Department of Medicine, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| | - Zacharie Drouin
- Division of Rheumatology, Department of Medicine, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| | - Nathalie Carrier
- Division of Rheumatology, Department of Medicine, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| | - Leopold Mbous Nguimbus
- Division of Rheumatology, Department of Medicine, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| | - Marie-Claude Beaulieu
- Department of Family and Emergency Medicine, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| | - Gilles Boire
- Division of Rheumatology, Department of Medicine, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| | - Alain Piché
- Division of Infectious Diseases, Department of Medicine, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| | - Hugues Allard-Chamard
- Division of Rheumatology, Department of Medicine, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| | - Sophie Roux
- Division of Rheumatology, Department of Medicine, Centre de Recherche du Centre Hospitalier de l’Université de Sherbrooke, 3001, 12th avenue N, Sherbrooke, PQ, Canada, J1H5N4
| |
Collapse
|
13
|
Dudley HM, O'Mara M, Auma A, Gong J, Ross Y, Gurevich N, Carbone S, Reihs A, Nguyen Y, McComsey GA, Cao Y, Balazs AB, Gordesky L, Payne M, Singer N, Kostadinova L, Wilson B, Zidar DA, King CL, Canaday DH, Shive CL, Mattar MM, Anthony DD. Rheumatoid arthritis and older age are associated with lower humoral and cellular immune response to primary series COVID-19 mRNA vaccine. Vaccine 2023; 41:6112-6119. [PMID: 37659895 PMCID: PMC11606055 DOI: 10.1016/j.vaccine.2023.08.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 09/04/2023]
Abstract
OBJECTIVE People with autoimmune disease have worse COVID-19 infection-related outcomes, lower antibody responses to COVID-19 vaccine, and higher rates of breakthrough infection. Immunosuppressive medications used to treat rheumatoid arthritis (RA) are associated with lower COVID-19 vaccine responses, though independent contributions of comorbidities, T-cell immunity, and age are less clear. We sought to test the hypothesis that RA, immunosuppressive medications used to treat RA, and older age, contribute to reduced B and T cell response to COVID-19 vaccine. METHODS We evaluated serum samples, taken the day of 1st vaccine dose, the day of 2nd dose, 2-6 weeks after 2nd dose, 7-12 weeks after 2nd dose, 13-24 weeks after 2nd dose, and 2-6 weeks after the 3rd dose, for anti-spike IgG and neutralizing antibody levels to Wuhan and Omicron BA.1 and peripheral blood mononuclear cells (PBMC) for spike-specific IFN-γ and IL-2 production by ELISPOT assay in 46 RA and 101 non-autoimmune control participants before and after the primary series COVID-19 mRNA vaccination. RESULTS RA participants had lower spike-specific IgG and Wuhan-strain neutralizing antibody levels 2-6 weeks compared to controls after the second dose of primary vaccine series. Neutralizing antibody levels against Omicron BA.1 were low in both groups. IFN-γ production correlated with Wuhan neutralizing antibody levels, while older age negatively correlated with spike-specific IL-2, IFN-γ and IgG. Lower antibody levels were associated with older age, RA status, and medication usage, while lower T cell responses were associated primarily with older age. CONCLUSIONS These data indicate lower COVID-19 mRNA vaccine-induced antibody levels in persons with RA compared to individuals without RA, likely partially attributable to immune suppressive medications. At the same time, older age is associated with lower antibody and cellular immune response to COVID-19 vaccines.
Collapse
Affiliation(s)
- Holly M Dudley
- Department of Molecular Biology, Case Western Reserve University School of Medicine, Cleveland, OH, United States; Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States; The MetroHealth System, Cleveland, OH, United States
| | - Megan O'Mara
- Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Ann Auma
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Jenny Gong
- Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States; The MetroHealth System, Cleveland, OH, United States
| | - Yael Ross
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States; The MetroHealth System, Cleveland, OH, United States
| | - Natalie Gurevich
- Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Sarah Carbone
- Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Alex Reihs
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Ynez Nguyen
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Grace A McComsey
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Yi Cao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | | | - Larraine Gordesky
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States; The MetroHealth System, Cleveland, OH, United States
| | - Michael Payne
- Department of Global Health, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Nora Singer
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States; The MetroHealth System, Cleveland, OH, United States
| | - Lenche Kostadinova
- Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Brigid Wilson
- Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - David A Zidar
- Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States; Department of Global Health, Case Western Reserve University School of Medicine, Cleveland, OH, United States; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Christopher L King
- Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States; Department of Global Health, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - David H Canaday
- Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Carey L Shive
- Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Maya M Mattar
- Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Donald D Anthony
- Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States; The MetroHealth System, Cleveland, OH, United States
| |
Collapse
|
14
|
Klebanoff SD, Rodda LB, Morishima C, Wener MH, Yuzefpolskiy Y, Bettelli E, Buckner JH, Speake C, Pepper M, Campbell DJ. Diminished responses to mRNA-based SARS-CoV-2 vaccines in individuals with rheumatoid arthritis on immune-modifying therapies. JCI Insight 2023; 8:e168663. [PMID: 37338983 PMCID: PMC10445680 DOI: 10.1172/jci.insight.168663] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/15/2023] [Indexed: 06/21/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disorder that causes debilitating swelling and destruction of the joints. People with RA are treated with drugs that actively suppress one or more parts of their immune system, and these may alter the response to vaccination against SARS-CoV-2. In this study, we analyzed blood samples from a cohort of patients with RA after receiving a 2-dose mRNA COVID-19 vaccine regimen. Our data show that individuals on the cytotoxic T lymphocyte antigen 4-Ig therapy abatacept had reduced levels of SARS-CoV-2-neutralizing antibodies after vaccination. At the cellular level, these patients showed reduced activation and class switching of SARS-CoV-2-specific B cells, as well as reduced numbers and impaired helper cytokine production by SARS-CoV-2-specific CD4+ T cells. Individuals on methotrexate showed similar but less severe defects in vaccine response, whereas individuals on the B cell-depleting therapy rituximab had a near-total loss of antibody production after vaccination. These data define a specific cellular phenotype associated with impaired response to SARS-CoV-2 vaccination in patients with RA on different immune-modifying therapies and help inform efforts to improve vaccination strategies in this vulnerable population.
Collapse
Affiliation(s)
- Samuel D. Klebanoff
- Benaroya Research Institute, Seattle, Washington, USA
- Department of Immunology and
| | | | - Chihiro Morishima
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Mark H. Wener
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | | | - Estelle Bettelli
- Benaroya Research Institute, Seattle, Washington, USA
- Department of Immunology and
| | - Jane H. Buckner
- Benaroya Research Institute, Seattle, Washington, USA
- Department of Immunology and
| | - Cate Speake
- Benaroya Research Institute, Seattle, Washington, USA
| | | | - Daniel J. Campbell
- Benaroya Research Institute, Seattle, Washington, USA
- Department of Immunology and
| |
Collapse
|
15
|
Petrone L, Sette A, de Vries RD, Goletti D. The Importance of Measuring SARS-CoV-2-Specific T-Cell Responses in an Ongoing Pandemic. Pathogens 2023; 12:862. [PMID: 37513709 PMCID: PMC10385870 DOI: 10.3390/pathogens12070862] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Neutralizing antibodies are considered a correlate of protection against SARS-CoV-2 infection and severe COVID-19, although they are not the only contributing factor to immunity: T-cell responses are considered important in protecting against severe COVID-19 and contributing to the success of vaccination effort. T-cell responses after vaccination largely mirror those of natural infection in magnitude and functional capacity, but not in breadth, as T-cells induced by vaccination exclusively target the surface spike glycoprotein. T-cell responses offer a long-lived line of defense and, unlike humoral responses, largely retain reactivity against the SARS-CoV-2 variants. Given the increasingly recognized role of T-cell responses in protection against severe COVID-19, the circulation of SARS-CoV-2 variants, and the potential implementation of novel vaccines, it becomes imperative to continuously monitor T-cell responses. In addition to "classical" T-cell assays requiring the isolation of peripheral blood mononuclear cells, simple whole-blood-based interferon-γ release assays have a potential role in routine T-cell response monitoring. These assays could be particularly useful for immunocompromised people and other clinically vulnerable populations, where interactions between cellular and humoral immunity are complex. As we continue to live alongside COVID-19, the importance of considering immunity as a whole, incorporating both humoral and cellular responses, is crucial.
Collapse
Affiliation(s)
- Linda Petrone
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, 00149 Rome, Italy;
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA;
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Rory D. de Vries
- Department Viroscience, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands;
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, 00149 Rome, Italy;
| |
Collapse
|
16
|
Brisotto G, Montico M, Turetta M, Zanussi S, Cozzi MR, Vettori R, Boschian Boschin R, Vinante L, Matrone F, Revelant A, Palazzari E, Innocente R, Fanetti G, Gerratana L, Garutti M, Lisanti C, Bolzonello S, Nicoloso MS, Steffan A, Muraro E. Integration of Cellular and Humoral Immune Responses as an Immunomonitoring Tool for SARS-CoV-2 Vaccination in Healthy and Fragile Subjects. Viruses 2023; 15:1276. [PMID: 37376576 DOI: 10.3390/v15061276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
Cellular and humoral immunity are both required for SARS-CoV-2 infection recovery and vaccine efficacy. The factors affecting mRNA vaccination-induced immune responses, in healthy and fragile subjects, are still under investigation. Thus, we monitored the vaccine-induced cellular and humoral immunity in healthy subjects and cancer patients after vaccination to define whether a different antibody titer reflected similar rates of cellular immune responses and if cancer has an impact on vaccination efficacy. We found that higher titers of antibodies were associated with a higher probability of positive cellular immunity and that this greater immune response was correlated with an increased number of vaccination side effects. Moreover, active T-cell immunity after vaccination was associated with reduced antibody decay. The vaccine-induced cellular immunity appeared more likely in healthy subjects rather than in cancer patients. Lastly, after boosting, we observed a cellular immune conversion in 20% of subjects, and a strong correlation between pre- and post-boosting IFN-γ levels, while antibody levels did not display a similar association. Finally, our data suggested that integrating humoral and cellular immune responses could allow the identification of SARS-CoV-2 vaccine responders and that T-cell responses seem more stable over time compared to antibodies, especially in cancer patients.
Collapse
Affiliation(s)
- Giulia Brisotto
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Marcella Montico
- Clinical Trial Office, Scientific Direction, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Matteo Turetta
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Stefania Zanussi
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Maria Rita Cozzi
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Roberto Vettori
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Romina Boschian Boschin
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Lorenzo Vinante
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Fabio Matrone
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Alberto Revelant
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Elisa Palazzari
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Roberto Innocente
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Giuseppe Fanetti
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Lorenzo Gerratana
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Mattia Garutti
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Camilla Lisanti
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Silvia Bolzonello
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Milena Sabrina Nicoloso
- Molecular Oncology Unit, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Elena Muraro
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| |
Collapse
|
17
|
Grifoni A, Alonzi T, Alter G, Noonan DM, Landay AL, Albini A, Goletti D. Impact of aging on immunity in the context of COVID-19, HIV, and tuberculosis. Front Immunol 2023; 14:1146704. [PMID: 37292210 PMCID: PMC10246744 DOI: 10.3389/fimmu.2023.1146704] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/11/2023] [Indexed: 06/10/2023] Open
Abstract
Knowledge of aging biology needs to be expanded due to the continuously growing number of elderly people worldwide. Aging induces changes that affect all systems of the body. The risk of cardiovascular disease and cancer increases with age. In particular, the age-induced adaptation of the immune system causes a greater susceptibility to infections and contributes to the inability to control pathogen growth and immune-mediated tissue damage. Since the impact of aging on immune function, is still to be fully elucidated, this review addresses some of the recent understanding of age-related changes affecting key components of immunity. The emphasis is on immunosenescence and inflammaging that are impacted by common infectious diseases that are characterized by a high mortality, and includes COVID-19, HIV and tuberculosis.
Collapse
Affiliation(s)
- Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Tonino Alonzi
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, Rome, Italy
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard, Cambridge, MA, United States
| | - Douglas McClain Noonan
- Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Alan L. Landay
- Department of Internal Medicine, Rush Medical College, Chicago, IL, United States
| | | | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, Rome, Italy
| |
Collapse
|
18
|
Alicandro G, Orena BS, Rosazza C, Cariani L, Russo M, Zatelli M, Badolato R, Gramegna A, Blasi F, Daccò V. Humoral and cell-mediated immune responses to BNT162b2 vaccine against SARS-CoV-2 in people with cystic fibrosis. Vaccine 2023:S0264-410X(23)00590-X. [PMID: 37263872 DOI: 10.1016/j.vaccine.2023.05.041] [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: 03/23/2023] [Revised: 04/26/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023]
Abstract
People with cystic fibrosis (pwCF) were considered to be clinically vulnerable to COVID-19 and were therefore given priority in the vaccination campaign. Vaccines induced a humoral response in these patients that was comparable to the response observed among the general population. However, the role of the cell-mediated immune response in providing long-term protection against SARS-CoV-2 in pwCF has not yet been defined. In this study, humoral (antibody titre) and cell-mediated immune responses (interferon-γ release) to the BNT162b2 vaccine were measured at different time points, from around 6-8 months after the 2nd dose and up to 8 months after the 3rd dose, in 118 CF patients and 26 non-CF subjects. Subjects were sampled between November 2021 and September 2022 and followed-up for breakthrough infection through October 2022. pwCF mounted a cell-mediated response that was similar to that observed in non-CF subjects. Low antibody titres (<1st quartile) were associated with a higher risk of breakthrough infection (HR: 2.39, 95 % CI: 1.17-4.88), while there was no significant association with low INF-γ levels (<0.3 IU/mL) (HR: 1.38, 95 % CI: 0.64-2.99). Further studies are needed in subgroup of pwCF receiving immunosuppressive therapy, such as organ transplant recipients. This data is important for tailoring vaccination strategies for this clinically vulnerable population.
Collapse
Affiliation(s)
- G Alicandro
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Cystic Fibrosis Center, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - B S Orena
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Microbiology Unit, Milan, Italy
| | - C Rosazza
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Cystic Fibrosis Center, Milan, Italy
| | - L Cariani
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Microbiology Unit, Milan, Italy
| | - M Russo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Cystic Fibrosis Center, Milan, Italy
| | - M Zatelli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Microbiology Unit, Milan, Italy
| | - R Badolato
- Department of Pediatrics, ASST Spedali Civili, Brescia, Italy; Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - A Gramegna
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy; Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy
| | - F Blasi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy; Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy
| | - V Daccò
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Cystic Fibrosis Center, Milan, Italy.
| |
Collapse
|
19
|
Ruggieri S, Aiello A, Tortorella C, Navarra A, Vanini V, Meschi S, Lapa D, Haggiag S, Prosperini L, Cuzzi G, Salmi A, Quartuccio ME, Altera AMG, Garbuglia AR, Ascoli Bartoli T, Galgani S, Notari S, Agrati C, Puro V, Nicastri E, Gasperini C, Goletti D. Dynamic Evolution of Humoral and T-Cell Specific Immune Response to COVID-19 mRNA Vaccine in Patients with Multiple Sclerosis Followed until the Booster Dose. Int J Mol Sci 2023; 24:ijms24108525. [PMID: 37239872 DOI: 10.3390/ijms24108525] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
This study characterizes antibody and T-cell immune responses over time until the booster dose of COronaVIrus Disease 2019 (COVID-19) vaccines in patients with multiple sclerosis (PwMS) undergoing different disease-modifying treatments (DMTs). We prospectively enrolled 134 PwMS and 99 health care workers (HCWs) having completed the two-dose schedule of a COVID-19 mRNA vaccine within the last 2-4 weeks (T0) and followed them 24 weeks after the first dose (T1) and 4-6 weeks after the booster (T2). PwMS presented a significant reduction in the seroconversion rate and anti-receptor-binding domain (RBD)-Immunoglobulin (IgG) titers from T0 to T1 (p < 0.0001) and a significant increase from T1 to T2 (p < 0.0001). The booster dose in PwMS showed a good improvement in the serologic response, even greater than HCWs, as it promoted a significant five-fold increase of anti-RBD-IgG titers compared with T0 (p < 0.0001). Similarly, the T-cell response showed a significant 1.5- and 3.8-fold increase in PwMS at T2 compared with T0 (p = 0.013) and T1 (p < 0.0001), respectively, without significant modulation in the number of responders. Regardless of the time elapsed since vaccination, most ocrelizumab- (77.3%) and fingolimod-treated patients (93.3%) showed only a T-cell-specific or humoral-specific response, respectively. The booster dose reinforces humoral- and cell-mediated-specific immune responses and highlights specific DMT-induced immune frailties, suggesting the need for specifically tailored strategies for immune-compromised patients to provide primary prophylaxis, early SARS-CoV-2 detection and the timely management of COVID-19 antiviral treatments.
Collapse
Affiliation(s)
- Serena Ruggieri
- Department of Neurosciences, San Camillo Forlanini Hospital, 00152 Rome, Italy
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Alessandra Aiello
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Carla Tortorella
- Department of Neurosciences, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Assunta Navarra
- Clinical Epidemiology Unit, National Institute for Infectious Disease Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Valentina Vanini
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
- UOS Professioni Sanitarie Tecniche, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Silvia Meschi
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Daniele Lapa
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Shalom Haggiag
- Department of Neurosciences, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Luca Prosperini
- Department of Neurosciences, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Gilda Cuzzi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Andrea Salmi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | | | - Anna Maria Gerarda Altera
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Anna Rosa Garbuglia
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Tommaso Ascoli Bartoli
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Simonetta Galgani
- Department of Neurosciences, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Stefania Notari
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Chiara Agrati
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital, 00146 Rome, Italy
| | - Vincenzo Puro
- UOC Emerging Infections and Centro di Riferimento AIDS (CRAIDS), National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Emanuele Nicastri
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Claudio Gasperini
- Department of Neurosciences, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| |
Collapse
|
20
|
The safety and immunogenicity of inactivated COVID-19 vaccine in old pulmonary tuberculosis patients. Eur J Clin Microbiol Infect Dis 2023; 42:503-512. [PMID: 36849838 PMCID: PMC9970849 DOI: 10.1007/s10096-023-04566-0] [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: 09/10/2022] [Accepted: 02/07/2023] [Indexed: 03/01/2023]
Abstract
The immunogenicity and safety of vaccines against coronavirus disease 2019 (COVID-19) remain unknown in patients with a history of pulmonary tuberculosis (OPTB). Therefore, the safety and effectiveness of inactivated vaccines against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) were assessed in patients with a history of PTB. The study cohort included 106 healthy controls and 93 adult patients with OPTB who received a two-dose vaccination. The study period was 21 to 105 days. Concentrations of antibodies (Abs) against receptor-binding domain (RBD) IgG and SARS-CoV-2 neutralizing Abs (NAbs) were measured, in addition to the frequencies of SARS-CoV-2-specific B and a portion T cells. The incidence of adverse events was similar between the OPTB patients and healthy controls. No severe adverse events occurred. Concentrations of Abs against RBD-IgG and CoV-2 neutralizing Abs in addition to the frequencies of RBD-specific memory B cells proportions were lower in OPTB patients than the healthy controls (all, p < 0.05), while the frequencies of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4+) cells were higher (p = 0.023). There was no obvious correlation between age and blood concentrations of Abs against RBD-IgG and CoV-2 neutralizing Abs, while immune responses were similar in the fibrosis and calcification groups. The period of time following full-course vaccination and lymphocyte counts were associated to anti-RBD-IgG responses. Inactivated COVID-19 vaccinations were well tolerated in OPTB patients, although immunogenicity was limited in this population. This study has been registered at ClinicalTrials.gov (NCT05043246).
Collapse
|
21
|
Aiello A, Coppola A, Ruggieri S, Farroni C, Altera AMG, Salmi A, Vanini V, Cuzzi G, Petrone L, Meschi S, Lapa D, Bettini A, Haggiag S, Prosperini L, Galgani S, Quartuccio ME, Bevilacqua N, Garbuglia AR, Agrati C, Puro V, Tortorella C, Gasperini C, Nicastri E, Goletti D. Longitudinal characterisation of B and T-cell immune responses after the booster dose of COVID-19 mRNA-vaccine in people with multiple sclerosis using different disease-modifying therapies. J Neurol Neurosurg Psychiatry 2023; 94:290-299. [PMID: 36522154 PMCID: PMC10086471 DOI: 10.1136/jnnp-2022-330175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND The decline of humoral response to COVID-19 vaccine led to authorise a booster dose. Here, we characterised the kinetics of B-cell and T-cell immune responses in patients with multiple sclerosis (PwMS) after the booster dose. METHODS We enrolled 22 PwMS and 40 healthcare workers (HCWs) after 4-6 weeks from the booster dose (T3). Thirty HCWs and 19 PwMS were also recruited 6 months (T2) after the first dose. Antibody response was measured by anti-receptor-binding domain (RBD)-IgG detection, cell-mediated response by an interferon (IFN)-γ release assay (IGRA), Th1 cytokines and T-cell memory profile by flow cytometry. RESULTS Booster dose increased anti-RBD-IgG titers in fingolimod-treated, cladribine-treated and IFN-β-treated patients, but not in ocrelizumab-treated patients, although antibody titres were lower than HCWs. A higher number of fingolimod-treated patients seroconverted at T3. Differently, T-cell response evaluated by IGRA remained stable in PwMS independently of therapy. Spike-specific Th1-cytokine response was mainly CD4+ T-cell-mediated, and in PwMS was significantly reduced (p<0.0001) with impaired IL-2 production compared with HCWs at T3. In PwMS, total Th1 and IFN-γ CD4+ T-cell responders to spike protein were increased from T2 to T3.Compared with HCWs, PwMS presented a higher frequency of CD4+ and CD8+ terminally differentiated effector memory cells and of CD4+ effector memory (TEM) cells, independently of the stimulus suggesting the association of this phenotype with MS status. CD4+ and CD8+ TEM cell frequency was further increased at T3 compared with T2. CONCLUSIONS COVID-19 vaccine booster strengthens humoral and Th1-cell responses and increases TEM cells in PwMS.
Collapse
Affiliation(s)
- Alessandra Aiello
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Andrea Coppola
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Serena Ruggieri
- Department of Human Neurosciences, University of Rome La Sapienza, Rome, Italy.,Neuroimmunology Unit, Santa Lucia Foundation Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Chiara Farroni
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Anna Maria Gerarda Altera
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Andrea Salmi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Valentina Vanini
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy.,Unità Operativa Semplice (UOS) Professioni Sanitarie Tecniche, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Gilda Cuzzi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Linda Petrone
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Silvia Meschi
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Daniele Lapa
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Aurora Bettini
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Shalom Haggiag
- Department of Neurosciences, San Camillo Forlanini Hospital, Rome, Italy
| | - Luca Prosperini
- Department of Neurosciences, San Camillo Forlanini Hospital, Rome, Italy
| | - Simonetta Galgani
- Department of Neurosciences, San Camillo Forlanini Hospital, Rome, Italy
| | | | - Nazario Bevilacqua
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Anna Rosa Garbuglia
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Chiara Agrati
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy.,Department of Pediatric Hematology and Oncology, Bambino Gesu Pediatric Hospital, Rome, Italy
| | - Vincenzo Puro
- UOC Emerging Infections and Centro di Riferimento AIDS (CRAIDS), National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Carla Tortorella
- Department of Neurosciences, San Camillo Forlanini Hospital, Rome, Italy
| | - Claudio Gasperini
- Department of Neurosciences, San Camillo Forlanini Hospital, Rome, Italy
| | - Emanuele Nicastri
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani Institute for Hospitalization and Care Scientific, Rome, Italy
| |
Collapse
|
22
|
Wroński J, Jaszczyk B, Roszkowski L, Felis-Giemza A, Bonek K, Kornatka A, Plebańczyk M, Burakowski T, Lisowska B, Kwiatkowska B, Maśliński W, Wisłowska M, Massalska M, Kuca-Warnawin E, Ciechomska M. The Kinetics of Humoral and Cellular Responses after the Booster Dose of COVID-19 Vaccine in Inflammatory Arthritis Patients. Viruses 2023; 15:v15030620. [PMID: 36992329 PMCID: PMC10052973 DOI: 10.3390/v15030620] [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: 01/17/2023] [Revised: 02/09/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Impaired immunogenicity of COVID-19 vaccinations in inflammatory arthritis (IA) patients results in diminished immunity. However, optimal booster vaccination regimens are still unknown. Therefore, this study aimed to assess the kinetics of humoral and cellular responses in IA patients after the COVID-19 booster. In 29 IA patients and 16 healthy controls (HC), humoral responses (level of IgG antibodies) and cellular responses (IFN-γ production) were assessed before (T0), after 4 weeks (T1), and after more than 6 months (T2) from the booster vaccination with BNT162b2. IA patients, but not HC, showed lower anti-S-IgG concentration and IGRA fold change at T2 compared to T1 (p = 0.026 and p = 0.031). Furthermore, in IA patients the level of cellular response at T2 returned to the pre-booster level (T0). All immunomodulatory drugs, except IL-6 and IL-17 inhibitors for the humoral and IL-17 inhibitors for the cellular response, impaired the immunogenicity of the booster dose at T2. Our study showed impaired kinetics of both humoral and cellular responses after the booster dose of the COVID-19 vaccine in IA patients, which, in the case of cellular response, did not allow the vaccination effect to be maintained for more than 6 months. Repetitive vaccination with subsequent booster doses seems to be necessary for IA patients.
Collapse
Affiliation(s)
- Jakub Wroński
- Department of Rheumatology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
- Correspondence:
| | - Bożena Jaszczyk
- Department of Outpatient Clinics, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Leszek Roszkowski
- Department of Outpatient Clinics, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Anna Felis-Giemza
- Biologic Therapy Center, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Krzysztof Bonek
- Department of Rheumatology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Anna Kornatka
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Magdalena Plebańczyk
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Tomasz Burakowski
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Barbara Lisowska
- Department of Anesthesiology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Brygida Kwiatkowska
- Department of Early Arthritis, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Włodzimierz Maśliński
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Małgorzata Wisłowska
- Department of Rheumatology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Magdalena Massalska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Ewa Kuca-Warnawin
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| | - Marzena Ciechomska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland
| |
Collapse
|
23
|
The Third Dose of BNT162b2 COVID-19 Vaccine Does Not “Boost” Disease Flares and Adverse Events in Patients with Rheumatoid Arthritis. Biomedicines 2023; 11:biomedicines11030687. [PMID: 36979666 PMCID: PMC10045021 DOI: 10.3390/biomedicines11030687] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 02/26/2023] Open
Abstract
Data on the risk of adverse events (AEs) and disease flares in autoimmune rheumatic diseases (ARDs) after the third dose of COVID-19 vaccine are scarce. The aim of this multicenter, prospective study is to analyze the clinical and immunological safety of BNT162b2 vaccine in a cohort of rheumatoid arthritis (RA) patients followed-up from the first vaccine cycle to the third dose. The vaccine showed an overall good safety profile with no patient reporting serious AEs, and a low percentage of total AEs at both doses (40/78 (51.3%) and 13/47 (27.7%) patients after the second and third dose, respectively (p < 0.002). Flares were observed in 10.3% of patients after the end of the vaccination cycle and 12.8% after the third dose. Being vaccinated for influenza was inversely associated with the onset of AEs after the second dose, at both univariable (p = 0.013) and multivariable analysis (p = 0.027). This result could allow identification of a predictive factor of vaccine tolerance, if confirmed in larger patient populations. A higher disease activity at baseline was not associated with a higher incidence of AEs or disease flares. Effectiveness was excellent after the second dose, with only 1/78 (1.3%) mild breakthrough infection (BI) and worsened after the third dose, with 9/47 (19.2%) BI (p < 0.002), as a probable expression of the higher capacity of the Omicron variants to escape vaccine recognition.
Collapse
|
24
|
Associations of HLA Polymorphisms with Anti-SARS-CoV-2 Spike and Neutralizing Antibody Titers in Japanese Rheumatoid Arthritis Patients Vaccinated with BNT162b2. Vaccines (Basel) 2023; 11:vaccines11020404. [PMID: 36851281 PMCID: PMC9965868 DOI: 10.3390/vaccines11020404] [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: 12/27/2022] [Revised: 01/23/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes Coronavirus Disease 2019. Anti-SARS-CoV-2 spike (S) and neutralizing antibodies (Abs) are measured to evaluate the efficacy of vaccines. Human leukocyte antigen (HLA) may be associated with vaccine efficacy. Here, we investigated the association of HLA polymorphisms with the production of anti-SARS-CoV-2 S or neutralizing Abs in vaccinated rheumatoid arthritis (RA) patients in Japan. Genotyping of DRB1 and DQB1 was conducted in 87 Japanese RA patients vaccinated with BNT162b2. Associations of allele or haplotype carrier frequencies with anti-SARS-CoV-2 S or neutralizing Abs were examined. DRB1*12:01 was significantly positively associated with the production of S Ab (p = 0.0225, odds ratio [OR] 6.08, 95% confidence interval [CI] 1.32-28.03). The DQB1*03:01 allele carrier frequency tended to be higher in high responders of S Ab. Allele carrier frequencies of DRB1*15:01 (p = 0.0102, OR 9.26, 95% CI 1.65-52.01) and DQB1*06:02 (p = 0.0373, OR 7.00, 95% CI 1.18-41.36) were higher in responders of neutralizing Ab. Haplotype and two-locus analyses of DRB1 and DQB1 suggested that DRB1 alleles were the primary drivers of these associations. Logistic regression analysis showed associations of these alleles independent of clinical characteristics. Independent associations were found between HLA alleles and anti-SARS-CoV-2 Ab production by vaccinated RA patients.
Collapse
|
25
|
Wroński J, Jaszczyk B, Roszkowski L, Felis-Giemza A, Bonek K, Kornatka A, Plebańczyk M, Burakowski T, Lisowska B, Kwiatkowska B, Maśliński W, Wisłowska M, Massalska M, Ciechomska M, Kuca-Warnawin E. Humoral and cellular immunogenicity of COVID-19 booster dose vaccination in inflammatory arthritis patients. Front Immunol 2022; 13:1033804. [PMID: 36389719 PMCID: PMC9659732 DOI: 10.3389/fimmu.2022.1033804] [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: 08/31/2022] [Accepted: 10/10/2022] [Indexed: 08/07/2023] Open
Abstract
Introduction Previous studies have shown a reduction in the effectiveness of primary COVID-19 vaccination in patients with rheumatic diseases. However, limited data is available regarding the effectiveness of the COVID-19 vaccine booster dose, especially on cellular response. The study aimed to assess the humoral and cellular immunogenicity of a booster dose in patients with inflammatory arthritis (IA). Patients and methods 49 IA and 47 age and sex-matched healthy controls (HC) were included in a prospective cohort study. Both groups completed primary COVID-19 vaccination and after more than 180 days received a BNT162b2 booster shot. Humoral responses (level of IgG antibodies) and cellular responses (IFN-γ production) were assessed before and after 4 weeks from the booster dose of the vaccine. Results After the booster dose, all participants showed an increased humoral response, although significantly reduced antibody levels were observed in IA patients compared to HC (p=0.004). The cellular response was significantly lower both before (p<0.001) and after (p<0.001) the booster dose in IA patients as compared to HC. Among the immunomodulatory drugs, only biological and targeted synthetic drugs lowered the humoral response after booster vaccination. However, the cellular response was decreased after all immunomodulatory drugs except IL-17 inhibitors and sulfasalazine. Conclusion Our data indicate that patients with rheumatic diseases present lower humoral and cellular responses after the COVID-19 booster vaccine in comparison to HC. This may translate into a recommendation for subsequent booster doses of the COVID-19 vaccine for rheumatic patients.
Collapse
Affiliation(s)
- Jakub Wroński
- Department of Rheumatology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Bożena Jaszczyk
- Department of Outpatient Clinics, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Leszek Roszkowski
- Department of Outpatient Clinics, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Anna Felis-Giemza
- Biologic Therapy Center, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Krzysztof Bonek
- Department of Rheumatology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Anna Kornatka
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Magdalena Plebańczyk
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Tomasz Burakowski
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Barbara Lisowska
- Department of Anesthesiology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Brygida Kwiatkowska
- Department of Early Arthritis, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Włodzimierz Maśliński
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Małgorzata Wisłowska
- Department of Rheumatology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Magdalena Massalska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Marzena Ciechomska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Ewa Kuca-Warnawin
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| |
Collapse
|
26
|
Qian G, Wang X, Patel NJ, Kawano Y, Fu X, Cook CE, Vanni KM, Kowalski EN, Banasiak EP, Bade KJ, Srivatsan S, Williams ZK, Todd DJ, Weinblatt ME, Wallace ZS, Sparks JA. Outcomes with and without outpatient SARS-CoV-2 treatment for patients with COVID-19 and systemic autoimmune rheumatic diseases: A retrospective cohort study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.10.27.22281629. [PMID: 36324801 PMCID: PMC9628202 DOI: 10.1101/2022.10.27.22281629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Objective To investigate temporal trends, severe outcomes, and rebound among systemic autoimmune rheumatic disease (SARD) patients according to outpatient SARS-CoV-2 treatment. Methods We performed a retrospective cohort study investigating outpatient SARS-CoV-2 treatments among SARD patients at Mass General Brigham (23/Jan/2022-30/May/2022). We identified SARS-CoV-2 infection by positive PCR or antigen test (index date=first positive test) and SARDs using diagnosis codes and immunomodulator prescription. Outpatient treatments were confirmed by medical record review. The primary outcome was hospitalization or death within 30 days following the index date. COVID-19 rebound was defined as documentation of negative then newly-positive SARS-CoV-2 tests. The association of any vs. no outpatient treatment with hospitalization/death was assessed using multivariable logistic regression. Results We analyzed 704 SARD patients with COVID-19 (mean age 58.4 years, 76% female, 49% with rheumatoid arthritis). Treatment as outpatient increased over calendar time (p<0.001). A total of 426(61%) received outpatient treatment: 307(44%) with nirmatrelvir/ritonavir, 105(15%) with monoclonal antibodies, 5(0.7%) with molnupiravir, 3(0.4%) with outpatient remdesivir, and 6(0.9%) with combinations. There were 9/426 (2.1%) hospitalizations/deaths among those treated as outpatient compared to 49/278 (17.6%) among those with no outpatient treatment (adjusted odds ratio [aOR] 0.12, 0.05 to 0.25). 25/318 (8%) of patients who received oral outpatient treatment had documented COVID-19 rebound. Conclusion Outpatient treatment was strongly associated with lower odds of severe COVID-19 compared to no outpatient treatment. At least 8% of SARD patients experienced COVID-19 rebound. These findings highlight the importance of outpatient COVID-19 treatment for SARD patients and the need for further research on rebound. KEY MESSAGES What is already known on this topic? Previous studies suggest that monoclonal antibodies are an effective outpatient treatment option for patients at high-risk of severe COVID-19, including those with systemic autoimmune rheumatic diseases (SARDs).Nirmatrelvir/ritonavir and molnupiravir are recently-authorized effective oral outpatient SARS-CoV-2 treatment options, but clinical trials were performed among the general population, mostly among unvaccinated and prior to Omicron viral variants.Oral outpatient SARS-CoV-2 treatments may result in COVID-19 rebound, characterized by newly-positive COVID-19 testing and recurrent symptoms, but no studies have investigated rebound prevalence among SARD patients. What this study adds? This is one of the first studies investigating outpatient SARS-CoV-2 treatments among SARD patients that includes oral options and quantifies the prevalence of COVID-19 rebound.Outpatient treatment was associated with 88% reduced odds of severe COVID-19 compared to no treatment.At least 8% of SARDs receiving oral outpatient treatment experienced COVID-19 rebound. How this study might affect research practice or policy? These results should encourage clinicians to prescribe and SARD patients to seek prompt outpatient COVID-19 treatment.This research provides an early estimate of the prevalence of COVID-19 rebound after oral outpatient treatment to quantify this risk to clinicians and SARD patients and encourage future research.
Collapse
Affiliation(s)
- Grace Qian
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA (60 Fenwood Road, Boston, MA, 02115)
| | - Xiaosong Wang
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA (60 Fenwood Road, Boston, MA, 02115)
| | - Naomi J. Patel
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, USA (Rheumatology Associates, 55 Fruit Street, Boston, MA, 02114)
- Harvard Medical School, Boston, MA
| | - Yumeko Kawano
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA (60 Fenwood Road, Boston, MA, 02115)
- Harvard Medical School, Boston, MA
| | - Xiaoqing Fu
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, USA (Rheumatology Associates, 55 Fruit Street, Boston, MA, 02114)
| | - Claire E. Cook
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, USA (Rheumatology Associates, 55 Fruit Street, Boston, MA, 02114)
| | - Kathleen M.M. Vanni
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA (60 Fenwood Road, Boston, MA, 02115)
| | - Emily N. Kowalski
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA (60 Fenwood Road, Boston, MA, 02115)
| | - Emily P. Banasiak
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA (60 Fenwood Road, Boston, MA, 02115)
| | - Katarina J. Bade
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA (60 Fenwood Road, Boston, MA, 02115)
| | - Shruthi Srivatsan
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, USA (Rheumatology Associates, 55 Fruit Street, Boston, MA, 02114)
| | - Zachary K. Williams
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, USA (Rheumatology Associates, 55 Fruit Street, Boston, MA, 02114)
| | - Derrick J. Todd
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA (60 Fenwood Road, Boston, MA, 02115)
- Harvard Medical School, Boston, MA
| | - Michael E. Weinblatt
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA (60 Fenwood Road, Boston, MA, 02115)
- Harvard Medical School, Boston, MA
| | - Zachary S. Wallace
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, USA (Rheumatology Associates, 55 Fruit Street, Boston, MA, 02114)
- Harvard Medical School, Boston, MA
- Clinical Epidemiology Program, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA (The Mongan Institute, 100 Cambridge Street, Suite 1600, Boston, MA, 02114)
| | - Jeffrey A. Sparks
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA (60 Fenwood Road, Boston, MA, 02115)
- Harvard Medical School, Boston, MA
| |
Collapse
|
27
|
Huang Y, Shin JE, Xu AM, Yao C, Joung S, Wu M, Zhang R, Shin B, Foley J, Mahov SB, Modes ME, Ebinger JE, Driver M, Braun JG, Jefferies CA, Parimon T, Hayes C, Sobhani K, Merchant A, Gharib SA, Jordan SC, Cheng S, Goodridge HS, Chen P. Evidence of premature lymphocyte aging in people with low anti-spike antibody levels after BNT162b2 vaccination. iScience 2022; 25:105209. [PMID: 36188190 PMCID: PMC9510055 DOI: 10.1016/j.isci.2022.105209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/22/2022] [Accepted: 09/22/2022] [Indexed: 11/26/2022] Open
Abstract
SARS-CoV-2 vaccines have unquestionably blunted the overall impact of the COVID-19 pandemic, but host factors such as age, sex, obesity, and other co-morbidities can affect vaccine efficacy. We identified individuals in a relatively healthy population of healthcare workers (CORALE study cohort) who had unexpectedly low peak anti-spike receptor binding domain (S-RBD) antibody levels after receiving the BNT162b2 vaccine. Compared to matched controls, "low responders" had fewer spike-specific antibody-producing B cells after the second and third/booster doses. Moreover, their spike-specific T cell receptor (TCR) repertoire had less depth and their CD4+ and CD8+T cell responses to spike peptide stimulation were less robust. Single cell transcriptomic evaluation of peripheral blood mononuclear cells revealed activation of aging pathways in low responder B and CD4+T cells that could underlie their attenuated anti-S-RBD antibody production. Premature lymphocyte aging may therefore contribute to a less effective humoral response and could reduce vaccination efficacy.
Collapse
Affiliation(s)
- Yapei Huang
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Juliana E. Shin
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Research Division of Immunology in the Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Alexander M. Xu
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Hematology and Cellular Therapy, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Changfu Yao
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sandy Joung
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Min Wu
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ruan Zhang
- Comprehensive Transplant Center, Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Bongha Shin
- Comprehensive Transplant Center, Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Joslyn Foley
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Hematology and Cellular Therapy, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Simeon B. Mahov
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Hematology and Cellular Therapy, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Matthew E. Modes
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Joseph E. Ebinger
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Matthew Driver
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jonathan G. Braun
- Research Division of Immunology in the Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Caroline A. Jefferies
- Research Division of Immunology in the Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Tanyalak Parimon
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Chelsea Hayes
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kimia Sobhani
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Akil Merchant
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Hematology and Cellular Therapy, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sina A. Gharib
- Computational Medicine Core at Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA 98109, USA
| | - Stanley C. Jordan
- Comprehensive Transplant Center, Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Helen S. Goodridge
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Research Division of Immunology in the Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Peter Chen
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
28
|
Infantino M, Tsalouchos A, Russo E, Laudicina S, Grossi V, Lari B, Benucci M, Stacchini L, Amedei A, Casprini P, Villalta D, Dattolo PC, Manfredi M. Assessing T-Cell Immunity in Kidney Transplant Recipients with Absent Antibody Production after a 3rd Dose of the mRNA-1273 Vaccine. Int J Mol Sci 2022; 23:12333. [PMID: 36293190 PMCID: PMC9604095 DOI: 10.3390/ijms232012333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/30/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022] Open
Abstract
The vulnerable population of kidney transplant recipients (KTRs) are low responders to COVID-19 vaccines, so specific immune surveillance is needed. The interferon-gamma (IFN-γ) release assay (IGRA) is effective in assessing T cell-mediated immunity. We assessed SARS-CoV-2-directed T cell responses in KTRs with absent antibody production after a third dose of the mRNA-1273 vaccine, using two different IGRAs. A cohort of 57 KTRs, who were actively followed up, received a third dose of the mRNA-1273 vaccine. After the evaluation of humoral immunity to SARS-CoV-2, 14 seronegative patients were tested with two commercial IGRAs (SD Biosensor and Euroimmun). Out of 14 patients, one and three samples were positive by IGRAs with Euroimmun and SD Biosensor, respectively. The overall agreement between the two assays was 85.7% (κ = 0.444). In addition, multivariate linear regression analysis showed no statistically significant association between the IFN-γ concentration, and the independent variables analyzed (age, gender, years since transplant, total lymphocytes cells/mcl, CD3+ cells/mcl, CD3+ CD4+ cells/mcl, CD3+ CD8+ cells/mcl, CD19+ cells/mcl, CD3-CD16+CD56+ cells/mcl) (p > 0.01). In a vulnerable setting, assessing cellular immune response to complement the humoral response may be advantageous. Since the two commercial IGRAs showed a good agreement on negative samples, the three discordant samples highlight the need for further investigations.
Collapse
Affiliation(s)
- Maria Infantino
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, 50143 Florence, Italy
| | - Aris Tsalouchos
- Nephrology and Dialysis Unit Firenze II, Santa Maria Annunziata Hospital, 50139 Florence, Italy
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Selene Laudicina
- Nephrology and Dialysis Unit Firenze II, Santa Maria Annunziata Hospital, 50139 Florence, Italy
| | - Valentina Grossi
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, 50143 Florence, Italy
| | - Barbara Lari
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, 50143 Florence, Italy
| | - Maurizio Benucci
- Rheumatology Unit, S. Giovanni di Dio Hospital, 50143 Florence, Italy
| | - Lorenzo Stacchini
- Department of Health Science, University of Florence, 50134 Florence, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Patrizia Casprini
- Clinical Pathology Laboratory, S. Giovanni di Dio Hospital, 50143 Florence, Italy
| | - Danilo Villalta
- Immunology and Allergology Laboratory Unit, S-Maria degli Angeli Hospital, 33170 Pordenone, Italy
| | - Pietro Claudio Dattolo
- Nephrology and Dialysis Unit Firenze II, Santa Maria Annunziata Hospital, 50139 Florence, Italy
| | - Mariangela Manfredi
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, 50143 Florence, Italy
| |
Collapse
|
29
|
Nemeth D, Vago H, Tothfalusi L, Ulakcsai Z, Becker D, Szabo Z, Rojkovich B, Merkely B, Nagy G. Factors influencing the SARS-CoV-2 infection and vaccination induced immune response in rheumatoid arthritis. Front Immunol 2022; 13:960001. [PMID: 36311767 PMCID: PMC9596981 DOI: 10.3389/fimmu.2022.960001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Background To investigate the factors that have significant impact on the Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) infection and vaccination induced immune response in rheumatoid arthritis (RA). Methods Serological response was measured by quantifying anti-SARS-CoV-2 specific antibodies, while the cell-mediated response was measured by a whole-blood test quantifying the interferon (IFN)-γ response to different SARS-CoV-2-specific domains. Results We prospectively enrolled 109 RA patients and 43 healthy controls. The median time (IQR) between the confirmed infection or the last vaccination dose and the day when samples were taken (“sampling interval”) was 3.67 (2.03, 5.50) months in the RA group. Anti-Spike (anti-S) specific antibodies were detected in 94% of RA patients. Among the investigated patient related variables, age (p<0.004), sampling interval (p<0.001), the brand of the vaccine (p<0.001) and targeted RA therapy (TNF-inhibitor, IL-6 inhibitor, anti-CD20 therapy) had significant effect on the anti-S levels. After covariate adjustment TNF-inhibitor therapy decreased the anti-S antibody concentrations by 80% (p<0.001). The same figures for IL-6 inhibitor and anti-CD20 therapy were 74% (p=0.049) and 97% (p=0.002), respectively. Compared to subjects who were infected but were not vaccinated, the RNA COVID-19 vaccines increased the anti-S antibody levels to 71.1 (mRNA-1273) and 36.0 (BNT162b2) fold (p<0.001). The corresponding figure for the ChAdOx1s vaccine is 18.1(p=0.037). Anti-CCP (anti-cyclic citrullinated peptides) positive patients had 6.28 times (p= 0.00165) higher anti-S levels, than the anti-CCP negative patients. Positive T-cell response was observed in 87% of the healthy volunteer group and in 52% of the RA patient group. Following vaccination or infection it declined significantly (p= 0.044) but more slowly than that of anti-S titer (6%/month versus 25%). Specific T-cell responses were decreased by 65% in patients treated with anti-CD20 therapy (p=0.055). Conclusion Our study showed that the SARS-CoV-2-specific antibody levels were substantially reduced in RA patients treated with TNF-α-inhibitors (N=51) and IL-6-inhibitor (N=15). In addition, anti-CD20 therapy (N=4) inhibited both SARS-CoV-2-induced humoral and cellular immune responses. Furthermore, the magnitude of humoral and cellular immune response was dependent on the age and decreased over time. The RNA vaccines and ChAdOx1s vaccine effectively increased the level of anti-S antibodies.
Collapse
Affiliation(s)
- Dora Nemeth
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- *Correspondence: Dora Nemeth,
| | - Hajnalka Vago
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Sports Medicine, Semmelweis University, Budapest, Hungary
| | - Laszlo Tothfalusi
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
| | | | - David Becker
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Zsofia Szabo
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
| | - Bernadett Rojkovich
- Buda Hospital of the Hospitaller Order of Saint John of God, Budapest, Hungary
| | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Sports Medicine, Semmelweis University, Budapest, Hungary
| | - Gyorgy Nagy
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| |
Collapse
|
30
|
Jolliffe DA, Faustini SE, Holt H, Perdek N, Maltby S, Talaei M, Greenig M, Vivaldi G, Tydeman F, Symons J, Davies GA, Lyons RA, Griffiths CJ, Kee F, Sheikh A, Shaheen SO, Richter AG, Martineau AR. Determinants of Antibody Responses to SARS-CoV-2 Vaccines: Population-Based Longitudinal Study (COVIDENCE UK). Vaccines (Basel) 2022; 10:1601. [PMID: 36298466 PMCID: PMC9610049 DOI: 10.3390/vaccines10101601] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/09/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Antibody responses to SARS-CoV-2 vaccines vary for reasons that remain poorly understood. A range of sociodemographic, behavioural, clinical, pharmacologic and nutritional factors could explain these differences. To investigate this hypothesis, we tested for presence of combined IgG, IgA and IgM (IgGAM) anti-Spike antibodies before and after 2 doses of ChAdOx1 nCoV-19 (ChAdOx1, AstraZeneca) or BNT162b2 (Pfizer-BioNTech) in UK adults participating in a population-based longitudinal study who received their first dose of vaccine between December 2020 and July 2021. Information on sixty-six potential sociodemographic, behavioural, clinical, pharmacologic and nutritional determinants of serological response to vaccination was captured using serial online questionnaires. We used logistic regression to estimate multivariable-adjusted odds ratios (aORs) for associations between independent variables and risk of seronegativity following two vaccine doses. Additionally, percentage differences in antibody titres between groups were estimated in the sub-set of participants who were seropositive post-vaccination using linear regression. Anti-spike antibodies were undetectable in 378/9101 (4.2%) participants at a median of 8.6 weeks post second vaccine dose. Increased risk of post-vaccination seronegativity associated with administration of ChAdOx1 vs. BNT162b2 (adjusted odds ratio (aOR) 6.6, 95% CI 4.2−10.4), shorter interval between vaccine doses (aOR 1.6, 1.2−2.1, 6−10 vs. >10 weeks), poor vs. excellent general health (aOR 3.1, 1.4−7.0), immunodeficiency (aOR 6.5, 2.5−16.6) and immunosuppressant use (aOR 3.7, 2.4−5.7). Odds of seronegativity were lower for participants who were SARS-CoV-2 seropositive pre-vaccination (aOR 0.2, 0.0−0.6) and for those taking vitamin D supplements (aOR 0.7, 0.5−0.9). Serologic responses to vaccination did not associate with time of day of vaccine administration, lifestyle factors including tobacco smoking, alcohol intake and sleep, or use of anti-pyretics for management of reactive symptoms after vaccination. In a sub-set of 8727 individuals who were seropositive post-vaccination, lower antibody titres associated with administration of ChAdOx1 vs. BNT162b2 (43.4% lower, 41.8−44.8), longer duration between second vaccine dose and sampling (12.7% lower, 8.2−16.9, for 9−16 weeks vs. 2−4 weeks), shorter interval between vaccine doses (10.4% lower, 3.7−16.7, for <6 weeks vs. >10 weeks), receiving a second vaccine dose in October−December vs. April−June (47.7% lower, 11.4−69.1), older age (3.3% lower per 10-year increase in age, 2.1−4.6), and hypertension (4.1% lower, 1.1−6.9). Higher antibody titres associated with South Asian ethnicity (16.2% higher, 3.0−31.1, vs. White ethnicity) or Mixed/Multiple/Other ethnicity (11.8% higher, 2.9−21.6, vs. White ethnicity), higher body mass index (BMI; 2.9% higher, 0.2−5.7, for BMI 25−30 vs. <25 kg/m2) and pre-vaccination seropositivity for SARS-CoV-2 (105.1% higher, 94.1−116.6, for those seropositive and experienced COVID-19 symptoms vs. those who were seronegative pre-vaccination). In conclusion, we identify multiple determinants of antibody responses to SARS-CoV-2 vaccines, many of which are modifiable.
Collapse
Affiliation(s)
- David A. Jolliffe
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AB, UK
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Sian E. Faustini
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Hayley Holt
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AB, UK
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
- Asthma UK Centre for Applied Research, Queen Mary University of London, London E1 2AB, UK
| | - Natalia Perdek
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Sheena Maltby
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Mohammad Talaei
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AB, UK
| | - Matthew Greenig
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Giulia Vivaldi
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AB, UK
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Florence Tydeman
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AB, UK
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | | | - Gwyneth A. Davies
- Population Data Science, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK
| | - Ronan A. Lyons
- Population Data Science, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK
| | - Christopher J. Griffiths
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AB, UK
- Asthma UK Centre for Applied Research, Queen Mary University of London, London E1 2AB, UK
| | - Frank Kee
- Centre for Public Health Research (NI), Queen’s University Belfast, Belfast BT12 6BA, UK
| | - Aziz Sheikh
- Usher Institute, University of Edinburgh, Edinburgh EH16 4UX, UK
| | - Seif O. Shaheen
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AB, UK
| | - Alex G. Richter
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Adrian R. Martineau
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AB, UK
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
- Asthma UK Centre for Applied Research, Queen Mary University of London, London E1 2AB, UK
| |
Collapse
|
31
|
Mrak D, Sieghart D, Simader E, Tobudic S, Radner H, Mandl P, Göschl L, Koblischke M, Hommer N, Wagner A, Mayer M, Schubert L, Hartl L, Kozbial K, Hofer P, Kartnig F, Hummel T, Kerschbaumer A, Deimel T, Puchner A, Gudipati V, Thalhammer R, Munda P, Uyanik-Ünal K, Zuckermann A, Novacek G, Reiberger T, Garner-Spitzer E, Reindl-Schwaighofer R, Kain R, Winkler S, Smolen JS, Stiasny K, Fischer GF, Perkmann T, Haslacher H, Zeitlinger M, Wiedermann U, Aberle JH, Aletaha D, Heinz LX, Bonelli M. Heterologous vector versus homologous mRNA COVID-19 booster vaccination in non-seroconverted immunosuppressed patients: a randomized controlled trial. Nat Commun 2022; 13:5362. [PMID: 36097029 PMCID: PMC9467419 DOI: 10.1038/s41467-022-33036-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 08/30/2022] [Indexed: 11/09/2022] Open
Abstract
Impaired response to COVID-19 vaccination is of particular concern in immunosuppressed patients. To determine the best vaccination strategy for this vulnerable group we performed a single center, 1:1 randomized blinded clinical trial. Patients who failed to seroconvert upon two mRNA vaccinations (BNT162b2 or mRNA-1273) are randomized to receive either a third dose of the same mRNA or the vector vaccine ChAdOx1 nCoV-19. Primary endpoint is the difference in SARS-CoV-2 spike antibody seroconversion rate between vector and mRNA vaccinated patients four weeks after the third dose. Secondary outcomes include cellular immune responses. Seroconversion rates at week four are significantly higher in the mRNA (homologous vaccination, 15/24, 63%) as compared to the vector vaccine group (heterologous vaccination, 4/22, 18%). SARS-CoV-2-specific T-cell responses are reduced but could be increased after a third dose of either vector or mRNA vaccine. In a multivariable logistic regression analysis, patient age and vaccine type are associated with seroconversion. No serious adverse event is attributed to COVID-19 booster vaccination. Efficacy and safety data underline the importance of a booster vaccination and support the use of a homologous mRNA booster vaccination in immunosuppressed patients.Trial registration: EudraCT No.: 2021-002693-10.
Collapse
Affiliation(s)
- Daniel Mrak
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Daniela Sieghart
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Simader
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Selma Tobudic
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Helga Radner
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Peter Mandl
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Lisa Göschl
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | | | - Nikolaus Hommer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Angelika Wagner
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, Vienna, Austria
| | - Margareta Mayer
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Lorenz Schubert
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Lukas Hartl
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Karin Kozbial
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Philipp Hofer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Felix Kartnig
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Hummel
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Deimel
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Antonia Puchner
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Venugopal Gudipati
- Institute of Hygiene and Applied Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Renate Thalhammer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Petra Munda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Keziban Uyanik-Ünal
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Andreas Zuckermann
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Gottfried Novacek
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Erika Garner-Spitzer
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, Vienna, Austria
| | - Roman Reindl-Schwaighofer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Renate Kain
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Stefan Winkler
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Josef S Smolen
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Gottfried F Fischer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, Vienna, Austria
| | - Judith H Aberle
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Leonhard X Heinz
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| | - Michael Bonelli
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
32
|
Aiello A, Coppola A, Vanini V, Petrone L, Cuzzi G, Salmi A, Altera AMG, Tortorella C, Gualano G, Gasperini C, Scolieri P, Beccacece A, Vita S, Bruzzese V, Lorenzetti R, Palmieri F, Nicastri E, Goletti D. Accuracy of QuantiFERON SARS-CoV-2 research use only assay and characterization of the CD4 + and CD8 + T cell-SARS-CoV-2 response: comparison with a homemade interferon-γ release assay. Int J Infect Dis 2022; 122:841-849. [PMID: 35878802 PMCID: PMC9307287 DOI: 10.1016/j.ijid.2022.07.049] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVES In this study, we aimed to characterize the SARS-CoV-2-specific T cell response detected by the QuantiFERON SARS-CoV-2 research use only assay in terms of accuracy and T cell subsets involved compared with a homemade interferon (IFN)-γ release assay (IGRA). METHODS We evaluated T cell response by the standardized QuantiFERON SARS-CoV-2 tubes (antigen [Ag]1 and Ag2) and a homemade IGRA quantifying IFN-γ response to SARS-CoV-2 spike peptides (homemade-IGRA-SPIKE test). We evaluated the T cell subsets mediating the specific response using flow cytometry. RESULTS We prospectively enrolled 66 individuals: COVID-19 or post-COVID-19 subjects and NO-COVID-19-vaccinated subjects, including healthy donors and immunocompromised subjects. The standardized kit detected 62.1% (41/66) of T cell responders. Ag2 tube showed a higher IFN-γ quantitative and qualitative response. Ag1 tube response was mainly mediated by CD4+ T cells; Ag2 tube response was mediated by CD4+ and CD8+ T cells. The homemade-IGRA-SPIKE test detected a higher number of responders (52/66, 78.8%) than the QuantiFERON SARS-CoV-2 assay (P = 0.056). The response was found in both T cell subsets, although a higher magnitude and response rate was observed in the CD4+ T cell subset. CONCLUSION The QuantiFERON SARS-CoV-2 response is mediated by CD4+ and CD8+ T cells. A lower number of responders is found compared with the homemade-IGRA-SPIKE test, likely because of the different peptide composition.
Collapse
Affiliation(s)
- Alessandra Aiello
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Andrea Coppola
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Valentina Vanini
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy,Unità Operativa Semplice (UOS) Professioni Sanitarie Tecniche, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Linda Petrone
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Gilda Cuzzi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Andrea Salmi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Anna Maria Gerarda Altera
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Carla Tortorella
- Department of Neurosciences, San Camillo Forlanini Hospital, Rome, Italy
| | - Gina Gualano
- Respiratory Infectious Diseases Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Claudio Gasperini
- Department of Neurosciences, San Camillo Forlanini Hospital, Rome, Italy
| | - Palma Scolieri
- UOC di Medicina e Rete Reumatologica, Nuovo Regina Margherita Hospital, Rome, Italy
| | - Alessia Beccacece
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Serena Vita
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Vincenzo Bruzzese
- UOC di Medicina e Rete Reumatologica, Nuovo Regina Margherita Hospital, Rome, Italy
| | - Roberto Lorenzetti
- UOC di Gastroenterologia ASL Roma1, Nuovo Regina Margherita, Rome, Italy
| | - Fabrizio Palmieri
- Respiratory Infectious Diseases Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Emanuele Nicastri
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy,Corresponding author: Translational Research Unit of the Research Department, National Institute for Infectious Diseases, Padiglione del Vecchio, Room 39, Via Portuense 292, Rome 00149, Italy. Tel.: +39 06 55170 906; fax: +39 06 5582 825
| |
Collapse
|
33
|
Furukawa H, Oka S, Higuchi T, Nakama M, Nagai N, Tohma S. Anti-SARS-CoV-2 Spike Antibody Titers and Neutralizing Antibodies in Vaccinated Rheumatoid Arthritis Patients. Vaccines (Basel) 2022; 10:vaccines10081365. [PMID: 36016252 PMCID: PMC9414304 DOI: 10.3390/vaccines10081365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A serological test is used to assess the efficacy of vaccination. It has been reported that anti-SARS-CoV-2 spike (S) and neutralizing antibody (Ab) levels are lower following vaccination in patients with rheumatic disease. Here, we investigated anti-SARS-CoV-2 S and neutralizing Abs in vaccinated rheumatoid arthritis (RA) patients in Japan. Anti-SARS-CoV-2 S and neutralizing Abs were quantified in 101 RA patients and 117 controls. Anti-SARS-CoV-2 S Ab levels were lower in RA patients than both earlier after vaccination in controls (mean RA 324.1 ± 591.8 SDM vs. control 1216.6 ± 854.4 [U/mL], p < 0.0001) and later after vaccination (324.1 ± 591.8 vs. 582.0 ± 415.6 [U/mL], p = 0.0002). The interval between vaccination of the RA patients and serum collection was longer than for controls early after vaccination (142.1 ± 31.6 vs. 98.3 ± 11.2 [days], p < 0.0001), but shorter than the later sample from the controls (142.1 ± 31.6 vs. 257.3 ± 11.2 [days], p < 0.0001). Importantly, anti-SARS-CoV-2 neutralizing Ab titers in RA patients were higher than in either early or later control samples (10.7 ± 4.9 vs. 8.6 ± 6.6 [%], p = 0.0072, and 10.7 ± 4.9 vs. 3.1 ± 3.7 [%], p < 0.0001, respectively). Anti-SARS-CoV-2 S Ab titers in vaccinated RA patients were lower than in controls, but they were influenced by other clinical manifestations. Anti-SARS-CoV-2 neutralizing Ab levels were independently increased in RA.
Collapse
Affiliation(s)
- Hiroshi Furukawa
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose 204-8585, Japan
- Correspondence: ; Tel.: +81-42-491-2111
| | - Shomi Oka
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose 204-8585, Japan
| | - Takashi Higuchi
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose 204-8585, Japan
- Department of Nephrology, Ushiku Aiwa General Hospital, 896 Shishiko-cho, Ushiku 300-1296, Japan
| | - Moriyuki Nakama
- Department of Clinical Laboratory, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose 204-8585, Japan
- Department of Clinical Laboratory, National Hospital Organization Shimofusa Psychiatric Medical Center, 578 Heta-cho, Midori-ku, Chiba 266-0007, Japan
| | - Nobuhiro Nagai
- Department of Clinical Laboratory, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose 204-8585, Japan
| | - Shigeto Tohma
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose 204-8585, Japan
| |
Collapse
|
34
|
Geisen UM, Rose R, Neumann F, Ciripoi M, Vullriede L, Reid HM, Berner DK, Bertoglio F, Hoff P, Hust M, Longardt AC, Lorentz T, Martini GR, Saggau C, Schirmer JH, Schubert M, Sümbül M, Tran F, Voß M, Zeuner R, Morrison PJ, Bacher P, Fickenscher H, Gerdes S, Peipp M, Schreiber S, Krumbholz A, Hoyer BF. The long term vaccine-induced anti-SARS-CoV-2 immune response is impaired in quantity and quality under TNFα blockade. J Med Virol 2022; 94:5780-5789. [PMID: 35945627 PMCID: PMC9538219 DOI: 10.1002/jmv.28063] [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: 07/07/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 01/06/2023]
Abstract
The humoral immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination in patients with chronic inflammatory disease (CID) declines more rapidly with tumor necrosis factor-α (TNF-α) inhibition. Furthermore, the efficacy of current vaccines against Omicron variants of concern (VOC) including BA.2 is limited. Alterations within immune cell populations, changes in IgG affinity, and the ability to neutralize a pre-VOC strain and the BA.2 virus were investigated in these at-risk patients. Serum levels of anti-SARS-CoV-2 IgG, IgG avidity, and neutralizing antibodies (NA) were determined in anti-TNF-α patients (n = 10) and controls (n = 24 healthy individuals; n = 12 patients under other disease-modifying antirheumatic drugs, oDMARD) before and after the second and third vaccination by ELISA, immunoblot and live virus neutralization assay. SARS-CoV-2-specific B- and T cell subsets were analysed by multicolor flow cytometry. Six months after the second vaccination, anti-SARS-CoV-2 IgG levels, IgG avidity and anti-pre-VOC NA titres were significantly reduced in anti-TNF-α recipients compared to controls (healthy individuals: avidity: p ≤ 0.0001; NA: p = 0.0347; oDMARDs: avidity: p = 0.0012; NA: p = 0.0293). The number of plasma cells was increased in anti-TNF-α patients (Healthy individuals: p = 0.0344; oDMARDs: p = 0.0254), while the absolute number of SARS-CoV-2-specific plasma cells 7 days after 2nd vaccination were comparable. Even after a third vaccination, these patients had lower anti-BA.2 NA titres compared to both other groups. We show a reduced SARS-CoV-2 neutralizing capacity in patients under TNF-α blockade. In this cohort, the plasma cell response appears to be less specific and shows stronger bystander activation. While these effects were observable after the first two vaccinations and with older VOC, the differences in responses to BA.2 were enhanced.
Collapse
Affiliation(s)
- Ulf Martin Geisen
- Medical Department I, Rheumatology and Clinical ImmunologyUniversity Medical Center Schleswig‐Holstein Campus KielKielGermany
| | - Ruben Rose
- Institute for Infection Medicine, Christian‐Albrecht University of Kiel and University Medical Center Schleswig‐HolsteinKielGermany
| | | | - Maria Ciripoi
- Medical Department I, Rheumatology and Clinical ImmunologyUniversity Medical Center Schleswig‐Holstein Campus KielKielGermany
| | - Lena Vullriede
- Medical Department I, Rheumatology and Clinical ImmunologyUniversity Medical Center Schleswig‐Holstein Campus KielKielGermany
| | - Hayley M. Reid
- Medical Department I, Rheumatology and Clinical ImmunologyUniversity Medical Center Schleswig‐Holstein Campus KielKielGermany
| | - Dennis Kristopher Berner
- Medical Department I, Rheumatology and Clinical ImmunologyUniversity Medical Center Schleswig‐Holstein Campus KielKielGermany
| | - Federico Bertoglio
- Department of BiotechnologyInstitute of Biochemistry, Biotechnology, and Bioinformatics, Technische Universität BraunschweigBraunschweigGermany
| | - Paula Hoff
- Department of Rheumatology, Endokrinologikum‐GruppeBerlinGermany
| | - Michael Hust
- Department of BiotechnologyInstitute of Biochemistry, Biotechnology, and Bioinformatics, Technische Universität BraunschweigBraunschweigGermany
| | | | | | - Gabriela Rios Martini
- Institute of ImmunologyUniversity Medical Center Schleswig‐HolsteinKielGermany,Department for DermatologyUniversity Medical Center Schleswig‐HolsteinKielGermany
| | - Carina Saggau
- Institute of ImmunologyUniversity Medical Center Schleswig‐HolsteinKielGermany
| | - Jan Henrik Schirmer
- Medical Department I, Rheumatology and Clinical ImmunologyUniversity Medical Center Schleswig‐Holstein Campus KielKielGermany
| | - Maren Schubert
- Department of BiotechnologyInstitute of Biochemistry, Biotechnology, and Bioinformatics, Technische Universität BraunschweigBraunschweigGermany
| | - Melike Sümbül
- Department for DermatologyUniversity Medical Center Schleswig‐HolsteinKielGermany
| | - Florian Tran
- Institute of Clinical Molecular BiologyChristian‐Albrecht University of KielKielGermany,Department for Internal Medicine IUniversity Medical Center Schleswig‐HolsteinKiel
| | - Mathias Voß
- Institute for Infection Medicine, Christian‐Albrecht University of Kiel and University Medical Center Schleswig‐HolsteinKielGermany
| | - Rainald Zeuner
- Medical Department I, Rheumatology and Clinical ImmunologyUniversity Medical Center Schleswig‐Holstein Campus KielKielGermany
| | - Peter J. Morrison
- Department for DermatologyUniversity Medical Center Schleswig‐HolsteinKielGermany
| | - Petra Bacher
- Institute of ImmunologyUniversity Medical Center Schleswig‐HolsteinKielGermany,Institute of Clinical Molecular BiologyChristian‐Albrecht University of KielKielGermany
| | - Helmut Fickenscher
- Institute for Infection Medicine, Christian‐Albrecht University of Kiel and University Medical Center Schleswig‐HolsteinKielGermany
| | - Sascha Gerdes
- Department for DermatologyUniversity Medical Center Schleswig‐HolsteinKielGermany
| | - Matthias Peipp
- Department of Internal Medicine II, Division of Antibody‐Based ImmunotherapyUniversity Medical Center Schleswig‐HolsteinKielGermany
| | - Stefan Schreiber
- Institute of Clinical Molecular BiologyChristian‐Albrecht University of KielKielGermany,Department for Internal Medicine IUniversity Medical Center Schleswig‐HolsteinKiel
| | - Andi Krumbholz
- Institute for Infection Medicine, Christian‐Albrecht University of Kiel and University Medical Center Schleswig‐HolsteinKielGermany,Labor Dr. Krause und Kollegen MVZ GmbHKielGermany
| | - Bimba Franziska Hoyer
- Medical Department I, Rheumatology and Clinical ImmunologyUniversity Medical Center Schleswig‐Holstein Campus KielKielGermany
| |
Collapse
|
35
|
Aiello A, Grossi A, Meschi S, Meledandri M, Vanini V, Petrone L, Casetti R, Cuzzi G, Salmi A, Altera AM, Pierelli L, Gualano G, Ascoli Bartoli T, Castilletti C, Agrati C, Girardi E, Palmieri F, Nicastri E, Di Rosa E, Goletti D. Coordinated innate and T-cell immune responses in mild COVID-19 patients from household contacts of COVID-19 cases during the first pandemic wave. Front Immunol 2022; 13:920227. [PMID: 35967321 PMCID: PMC9364317 DOI: 10.3389/fimmu.2022.920227] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/30/2022] [Indexed: 01/08/2023] Open
Abstract
Objective To better define the immunopathogenesis of COVID-19, the present study aims to characterize the early immune responses to SARS-CoV-2 infection in household contacts of COVID-19 cases. In particular, innate, T- and B-cell specific responses were evaluated over time. Methods Household contacts of COVID-19 cases screened for SARS−CoV−2 infection by nasopharyngeal swab for surveillance purposes were enrolled (T0, n=42). Of these, 28 subjects returned for a follow-up test (T1). The innate response was assessed by detecting a panel of soluble factors by multiplex-technology in plasma samples. Cell-mediated response was evaluated by measuring interferon (IFN)-γ levels by ELISA in plasma harvested from whole-blood stimulated with SARS−CoV−2 peptide pools, including spike (S), nucleocapsid (N) and membrane (M) proteins. The serological response was assessed by quantifying anti-Receptor-Binding-Domain (RBD), anti-Nucleocapsid (N), whole virus indirect immunofluorescence, and neutralizing antibodies. Results At T0, higher levels of plasmatic IFN-α, IL-1ra, MCP-1 and IP-10, and lower levels of IL-1β, IL-9, MIP-1β and RANTES were observed in subjects with positive swab compared to individuals with a negative one (p<0.05). Plasmatic IFN-α was the only cytokine detectable in subjects with positive SARS-CoV-2 swabs with high accuracy for swab score positivity (0.93, p<0.0001). Among subjects with positive swabs, significant negative correlations were found among the RT-PCR cycle threshold values reported for genes S and N and IFN-α or IP-10 levels. At T0, the IFN-γ T-cell specific response was detected in 50% (5/10) of subjects with positive swab, while anti-RBD/anti-N antibodies showed a positivity rate of 10% (1/10). At T1, the IFN-γ T-cell specific response was detected in most of the confirmed-infection subjects (77.8%, 7/9), whereas the serological response was still observed in a minority of them (44.4%, 4/9). Overall, the swab test showed a moderate concordance with the T-cell response (78.6%, k=0.467), and a scarce concordance with the serological one (72.9%, k=0.194). Conclusions Plasmatic IFN-α and the IFN-γ T-cell specific response appear early even in the absence of seroconversion, and show a greater positivity rate than the serological response in household contacts with positive swab.
Collapse
Affiliation(s)
- Alessandra Aiello
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Adriano Grossi
- Local Public Health Office, Azienda Sanitaria Locale (ASL) Roma 1, Rome, Italy
| | - Silvia Meschi
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Marcello Meledandri
- Unità Operativa Complessa (UOC) Microbiology and Virology, Azienda Sanitaria Locale (ASL) Roma 1-San Filippo Neri Hospital, Rome, Italy
| | - Valentina Vanini
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
- Unità Operativa Semplice (UOS) Professioni Sanitarie Tecniche, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Linda Petrone
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Rita Casetti
- Laboratory of Cellular Immunology, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Gilda Cuzzi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Andrea Salmi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Anna Maria Altera
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Luca Pierelli
- Unità Operativa Complessa (UOC) Transfusion Medicine and Stem Cell, San Camillo Forlanini Hospital, Rome, Italy
| | - Gina Gualano
- Respiratory Infectious Diseases Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Tommaso Ascoli Bartoli
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Concetta Castilletti
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Chiara Agrati
- Laboratory of Cellular Immunology, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Enrico Girardi
- Clinical Epidemiology, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Fabrizio Palmieri
- Respiratory Infectious Diseases Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Emanuele Nicastri
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Enrico Di Rosa
- Local Public Health Office, Azienda Sanitaria Locale (ASL) Roma 1, Rome, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
- *Correspondence: Delia Goletti,
| |
Collapse
|
36
|
Ferri C, Gragnani L, Raimondo V, Visentini M, Giuggioli D, Lorini S, Foti R, Cacciapaglia F, Caminiti M, Olivo D, Cuomo G, Pellegrini R, Pigatto E, Urraro T, Naclerio C, Tavoni A, Puccetti L, Cavazzana I, Ruscitti P, Vadacca M, La Gualana F, Cozzi F, Spinella A, Visalli E, Bosco YD, Amato G, Masini F, Mariano GP, Brittelli R, Aiello V, Scorpiniti D, Rechichi G, Varcasia G, Monti M, Elia G, Franceschini F, Casato M, Ursini F, Giacomelli R, Fallahi P, Santini SA, Iannone F, Salvarani C, Zignego AL, Antonelli A. Absent or suboptimal response to booster dose of COVID-19 vaccine in patients with autoimmune systemic diseases. J Autoimmun 2022; 131:102866. [PMID: 35841684 PMCID: PMC9271490 DOI: 10.1016/j.jaut.2022.102866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 11/25/2022]
Abstract
Autoimmune systemic diseases (ASD) show impaired immunogenicity to COVID-19 vaccines. Our prospective observational multicenter study aimed at evaluating the seroconversion elicited by COVID-19 vaccine over the entire vaccination cycle including the booster dose. Among 478 unselected ASD patients originally evaluated at the end of the first vaccination cycle (time 1), 344 individuals were re-evaluated after a 6-month period (time 2), and 244 after the booster vaccine dose (time 3). The immunogenicity of mRNA COVID-19 vaccines (BNT162b2 and mRNA-1273) was assessed by measuring serum IgG-neutralizing antibody (NAb) on samples obtained at the three time points in both patients and 502 age-matched controls. In the 244 ASD group that received booster vaccine and monitored over the entire follow-up, the mean serum NAb levels (time 1, 2, and 3: 696.8 ± 52.68, 370.8 ± 41.92, and 1527 ± 74.16SD BAU/mL, respectively; p < 0.0001) were constantly lower compared to controls (p < 0.0001), but they significantly increased after the booster dose compared to the first two measurements (p < 0.0001). The percentage of patients with absent/suboptimal response to vaccine significantly decreased after the booster dose compared to the first and second evaluations (time 1, 2, and 3: from 28.2% to 46.3%, and to 7.8%, respectively; p < 0.0001). Of note, the percentage of patients with absent/suboptimal response after the booster dose was significantly higher compared to controls (19/244, 7.8% vs 1/502, 0.2%; p < 0.0001). Similarly, treatment with immune-modifiers increased the percentage of patients exhibiting absent/suboptimal response (16/122, 13.1% vs 3/122, 2.46%; p = 0.0031). Overall, the above findings indicate the usefulness of booster vaccine administration in ASD patients. Moreover, the persistence of a significantly higher percentage of individuals without effective seroconversion (7.8%), even after the booster dose, warrants for careful monitoring of NAb levels in all ASD patients to identify those with increased risk of infection. In this particularly frail patients’ setting, tailored vaccination and/or therapeutic strategy are highly advisable.
Collapse
Affiliation(s)
- Clodoveo Ferri
- Rheumatology Unit, University of Modena and Reggio Emilia, School of Medicine, Modena, Italy; Rheumatology Clinic 'Madonna dello Scoglio' Cotronei, Crotone, Italy.
| | - Laura Gragnani
- MASVE Interdepartmental Hepatology Center, Department of Experimental and clinical Medicine, University of Florence Center, Center for Research and Innovation CRIA-MASVE, Firenze, Italy
| | - Vincenzo Raimondo
- Rheumatology Clinic 'Madonna dello Scoglio' Cotronei, Crotone, Italy
| | - Marcella Visentini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Dilia Giuggioli
- Rheumatology Unit, University of Modena and Reggio Emilia, School of Medicine, Modena, Italy
| | - Serena Lorini
- MASVE Interdepartmental Hepatology Center, Department of Experimental and clinical Medicine, University of Florence Center, Center for Research and Innovation CRIA-MASVE, Firenze, Italy
| | - Rosario Foti
- AOU Policlinico Vittorio Emanuele, Catania, Italy
| | | | - Maurizio Caminiti
- UOD Reumatologia- Grande Ospedale Metropolitano, Reggio Calabria, Italy
| | - Domenico Olivo
- Rheumatology Outpatient Clinic, San Giovanni di Dio Hospital, Crotone, Italy
| | | | | | | | - Teresa Urraro
- Rheumatology Unit, "M. Scarlato" Hospital, Scafati (SA), Italy
| | | | | | | | | | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological & Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marta Vadacca
- Unità Operativa di Immunoreumatologia-Area Medicina Clinica Policlinico Universitario Campus Bio-Medico di Roma, Roma, Italy
| | - Francesca La Gualana
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Amelia Spinella
- Rheumatology Unit, University of Modena and Reggio Emilia, School of Medicine, Modena, Italy
| | | | | | | | | | | | | | - Vincenzo Aiello
- Rheumatology Clinic 'Madonna dello Scoglio' Cotronei, Crotone, Italy
| | | | - Giovanni Rechichi
- Rheumatology Clinic 'Madonna dello Scoglio' Cotronei, Crotone, Italy
| | | | - Monica Monti
- MASVE Interdepartmental Hepatology Center, Department of Experimental and clinical Medicine, University of Florence Center, Center for Research and Innovation CRIA-MASVE, Firenze, Italy
| | - Giusy Elia
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, School of Medicine, Pisa, Italy
| | | | - Milvia Casato
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesco Ursini
- Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Roberto Giacomelli
- Unità Operativa di Immunoreumatologia-Area Medicina Clinica Policlinico Universitario Campus Bio-Medico di Roma, Roma, Italy
| | - Poupak Fallahi
- Department of Translational Research & New Technologies in Medicine and Surgery, University of Pisa, School of Medicine, Pisa, Italy
| | - Stefano Angelo Santini
- Department of Basic, Clinical, Intensive and Perioperative Biotechnological Sciences, Catholic University School of Medicine, Rome, Italy; Synlab Italia, Monza (MB), Italy
| | | | - Carlo Salvarani
- Rheumatology Unit, University of Modena and Reggio Emilia, School of Medicine, Modena, Italy
| | - Anna Linda Zignego
- MASVE Interdepartmental Hepatology Center, Department of Experimental and clinical Medicine, University of Florence Center, Center for Research and Innovation CRIA-MASVE, Firenze, Italy
| | - Alessandro Antonelli
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, School of Medicine, Pisa, Italy
| |
Collapse
|
37
|
Blank RB, Haberman RH, Qian K, Samanovic M, Castillo R, Jimenez Hernandez A, Vasudevapillai Girija P, Catron S, Uddin Z, Rackoff P, Solomon G, Azar N, Rosenthal P, Izmirly P, Samuels J, Golden B, Reddy S, Mulligan MJ, Hu J, Scher JU. Low incidence and transient elevation of autoantibodies post mRNA COVID-19 vaccination in inflammatory arthritis. Rheumatology (Oxford) 2022; 62:467-472. [PMID: 35640110 PMCID: PMC9213868 DOI: 10.1093/rheumatology/keac322] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/06/2022] [Accepted: 05/24/2022] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES Autoantibody seroconversion has been extensively studied in the context of COVID-19 infection but data regarding post-vaccination autoantibody production is lacking. Here we aimed to determine the incidence of common autoantibody formation following mRNA COVID-19 vaccines in patients with inflammatory arthritis (IA) and in healthy controls. METHODS Autoantibody seroconversion was measured by serum ELISA in a longitudinal cohort of IA participants and healthy controls before and after COVID-19 mRNA-based immunization. RESULTS Overall, there was a significantly lower incidence of ANA seroconversion in participants who did not contract COVID-19 prior to vaccination compared with those who been previously infected (7.4% vs 24.1%, P = 0.014). Incidence of de novo anti-CCP seroconversion in all participants was low at 4.9%. Autoantibody levels were typically of low titre, transient, and not associated with increase in IA flares. CONCLUSIONS In both health and inflammatory arthritis, the risk of autoantibody seroconversion is lower following mRNA-based immunization than following natural SARS-CoV-2 infection. Importantly, seroconversion does not correlate with self-reported IA disease flare risk, further supporting the encouragement of mRNA-based COVID-19 immunization in the IA population.
Collapse
Affiliation(s)
- Rebecca B Blank
- Correspondence to: Rebecca B. Blank, Division of Rheumatology, New York University School of Medicine, 301 East 17th St, Suite 1400, New York, NY 10003, USA. E-mail:
| | | | - Kun Qian
- Division of Biostatistics, Department of Population Health
| | - Marie Samanovic
- NYU Langone Vaccine Center, NYU School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Mark J Mulligan
- NYU Langone Vaccine Center, NYU School of Medicine, New York, NY, USA
| | - Jiyuan Hu
- Division of Biostatistics, Department of Population Health
| | | |
Collapse
|
38
|
COVID-19 vaccine immunogenicity in 16 patients with autoimmune systemic diseases. Lack of both humoral and cellular response to booster dose and ongoing disease modifying therapies. J Transl Autoimmun 2022; 5:100164. [PMID: 36120415 PMCID: PMC9472465 DOI: 10.1016/j.jtauto.2022.100164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/10/2022] [Accepted: 09/12/2022] [Indexed: 01/14/2023] Open
Abstract
Background Patients with autoimmune systemic diseases (ASDs) represent a frail population during the ongoing COVID-19 pandemic. The vaccination is the major preventive measure; however, a significant number of ASD patients show an impaired production of anti-COVID-19 neutralizing antibodies (NAb), possibly counterbalanced by adequate T-cell response. The present study aimed at evaluating both humoral and cellular response to COVID-19 vaccine booster dose in this particular setting. Patients and methods Serum NAb titer and T-cell response (measuring interferon gamma –IFN–γ- release) were evaluated 3 weeks after the COVID-19 vaccine booster dose, in 17 patients (12 F, mean age 68.8 ± 15.3 SD yrs) with different ASDs, compared to 17 healthy controls (HCs). Results The analysis excluded one patient reporting symptoms of COVID-19 only after the immunogenicity tests had been performed. The NAb levels were significantly lower in ASD compared to HCs (p < 0.0001); moreover, patients showed a higher percentage of negative/sub-optimal humoral response (31% vs 0% of HCs; p = 0.0184). The study of cellular response showed lower levels of IFN-γ for both Ag1 (p = 0.0032) and Ag2 (p = 0.0136) in ASD patients compared to HCs, as well lower rate of adequate T-cell response compared to HCs (50% vs 94%; p = 0.0066). Disease modifying therapies (DMT) were administered in all patients with deficient NAb production (5/5, 100%), but in only 3/11 (27%) of responders (p = 0.025). Worthy to note, 3/16 (19%) ASD patients developed neither humoral nor cellular responses, all treated with DMT. Conclusions The impaired immunogenicity to COVID-19 vaccine booster and even more the concomitant lack of both humoral and cellular response might represent a high risk for severe COVID-19, particularly in ASD patients undergoing DMT. These frail subjects should be tightly monitored for their immune protection and prioritized for the fourth dose of COVID-19 vaccine. Moreover, in the occurrence of SARS-CoV2 infection, treatments with specific monoclonal antibodies and/or antivirals may be highly recommendable.
Collapse
|