1
|
Kamegai K, Itoh N, Ishikane M, Iwamoto N, Asai Y, Akazawa-Kai N, Fuwa N, Takasaki J, Hojo M, Hangaishi A, Togano T, Teruya K, Takahashi K, Miyamoto S, Hirata Y, Kanno T, Saito T, Katano H, Suzuki T, Ohmagari N. Duration of infectious virus shedding of SARS-CoV-2 Omicron variant among immunocompromised patients. J Infect Chemother 2025; 31:102631. [PMID: 39848541 DOI: 10.1016/j.jiac.2025.102631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/06/2025] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
OBJECTIVE The duration of viral shedding and criteria for de-isolation in the hospital among immunocompromised patients with coronavirus disease 2019 (COVID-19) remain unclear. This study aimed to evaluate viral shedding duration in immunocompromised patients infected with the Omicron variant of severe acute respiratory syndrome coronavirus 2. METHODS A prospective cohort study was performed at 2 tertiary medical centers in Japan during the Omicron epidemic waves from July 2022 to January 2023. Nasopharyngeal swabs were serially collected from immunocompromised patients with COVID-19, including those with hematological malignancies, solid tumors, autoimmune diseases, and human immunodeficiency virus infection. Patients were classified as severely or moderately immunocompromised according to the Japanese national guidelines for tixagevimab-cilgavimab. The relationship between patient characteristics, immune status, duration of viral RNA presence, and infectious virus shedding were assessed using Mann-Whitney U and Fisher's exact tests. RESULTS Among 41 patients (163 samples), 9 (47 samples) were severely and 32 (116 samples) were moderately immunocompromised. In the severely and moderately immunocompromised groups, 87.2 % and 75.0 % of the samples were viral RNA-positive, while 36.2 % and 35.3 % were culture-positive, respectively. Five culture-positive samples after day 20 were from 2 severely immunocompromised patients on B cell depletion therapy. No culture-positive samples were found for the moderately immunocompromised patients after day 10. CONCLUSIONS Long-term viral shedding should be closely monitored in severely immunocompromised patients with COVID-19.
Collapse
Affiliation(s)
- Kohei Kamegai
- Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, 162-8655, Tokyo, Japan
| | - Naoya Itoh
- Division of Infectious Diseases, Aichi Cancer Center Hospital, Aichi, Japan; Department of Infectious Diseases, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan; Nagoya City University East Medical Center, Aichi, Japan; Department of Clinical Infectious Diseases, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Masahiro Ishikane
- Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, 162-8655, Tokyo, Japan.
| | - Noriko Iwamoto
- Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, 162-8655, Tokyo, Japan
| | - Yusuke Asai
- Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, 162-8655, Tokyo, Japan
| | - Nana Akazawa-Kai
- Division of Infectious Diseases, Aichi Cancer Center Hospital, Aichi, Japan; Department of Infectious Diseases, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan; Nagoya City University East Medical Center, Aichi, Japan
| | - Noriko Fuwa
- Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, 162-8655, Tokyo, Japan
| | - Jin Takasaki
- Department of Pulmonology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Masayuki Hojo
- Department of Pulmonology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Akira Hangaishi
- Department of Hematology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tomiteru Togano
- Department of Hematology, National Center for Global Health and Medicine, Tokyo, Japan; Division of Hematology, Shonan Kamakura General Hospital, Kanagawa, Japan
| | - Katsuji Teruya
- AIDS Clinical Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kenichiro Takahashi
- Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Tokyo, Japan
| | - Sho Miyamoto
- Department of Pathology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, 162-8640, Tokyo, Japan
| | - Yuichiro Hirata
- Department of Pathology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, 162-8640, Tokyo, Japan
| | - Takayuki Kanno
- Department of Pathology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, 162-8640, Tokyo, Japan
| | - Tomoya Saito
- Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Tokyo, Japan
| | - Harutaka Katano
- Department of Pathology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, 162-8640, Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, 162-8640, Tokyo, Japan.
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, 162-8655, Tokyo, Japan
| |
Collapse
|
2
|
Canziani LM, Azzini AM, Salmanton-García J, Savoldi A, Caponcello MG, Pasquini Z, Pagano L, Cornely OA, Cingolani A, Mazzotta V, Cosentino F, Baño JR, Krampera M, Tacconelli E. ORCHESTRA Delphi consensus: diagnostic and therapeutic management of SARS-CoV-2 infection in haematological patients. Clin Microbiol Infect 2025:S1198-743X(25)00122-3. [PMID: 40122205 DOI: 10.1016/j.cmi.2025.03.004] [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: 12/19/2024] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVES COVID-19 poses a significant risk to individuals with haematological malignancies (HM), as they are particularly vulnerable to severe disease progression and hospitalization due to their compromised immune systems. Many clinical decisions regarding the management of COVID-19 in these patients are yet to be fully addressed by existing guidelines, leading to variability in care. METHODS A 28-item Delphi survey was developed to gather expert opinions on key areas of COVID-19 management in patients with HM, including risk stratification for severe COVID-19, diagnostic processes, and treatment decisions. RESULTS Twenty-one experts with backgrounds in haematology and infectious diseases were enrolled. Of the 28 questions posed to the experts, consensus was reached on 15 statements. DISCUSSION These Delphi consensus statements offer valuable suggestions with direct implications for clinical practice, addressing critical areas such as risk identification, appropriate diagnostic approaches, and tailored treatment strategies for patients with HM with COVID-19. The findings provide actionable insights that may help fill gaps in current scientific literature, enhancing patient care and decision-making in this high-risk population.
Collapse
Affiliation(s)
- Lorenzo Maria Canziani
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy.
| | - Anna Maria Azzini
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Jon Salmanton-García
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University Hospital Cologne, Cologne, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf and Excellence Center for Medical Mycology, Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Alessia Savoldi
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Maria Giulia Caponcello
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen, Macarena, Seville, Spain; Departamento de Medicina, Universidad de Sevilla, Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Seville, Spain; CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Zeno Pasquini
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Livio Pagano
- Hematology Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS-Università Cattolica del Sacro Cuore, Rome, Italy
| | - Oliver A Cornely
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University Hospital Cologne, Cologne, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf and Excellence Center for Medical Mycology, Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany; Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, Cologne, Germany
| | - Antonella Cingolani
- Infectious Diseases Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS-Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Mazzotta
- Clinical Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Federica Cosentino
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, ARNAS Garibaldi Nesima Hospital, University of Catania, Catania, Italy
| | - Jesús Rodríguez Baño
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen, Macarena, Seville, Spain; Departamento de Medicina, Universidad de Sevilla, Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Seville, Spain; CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Mauro Krampera
- Hematology and Bone Marrow Transplant Unit, Section of Biomedicine of Innovation, Department of Engineering for Innovative Medicine, University of Verona, Verona, Italy
| | - Evelina Tacconelli
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| |
Collapse
|
3
|
Patel NJ, Srivatsan S, Kowalski EN, King A, Wang X, Vanni KM, Qian G, Hanberg JS, Bade KJ, Saavedra AA, Mueller KT, Hang B, Williams ZK, Johnson C, Negron M, Sparks JA, Wallace ZS. Patients with systemic autoimmune rheumatic diseases remain at risk for hospitalisation for COVID-19 infection in the Omicron era (2022-2024): a retrospective cohort study. RMD Open 2025; 11:e005114. [PMID: 40044572 PMCID: PMC11883534 DOI: 10.1136/rmdopen-2024-005114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
OBJECTIVE To investigate the risk factors for severe acute COVID-19 outcomes in the Omicron era among individuals with systemic autoimmune rheumatic diseases (SARDs). METHODS We identified patients with confirmed SARDs and COVID-19 (positive PCR and/or antigen test) from 1 September 2022 to 15 March 2024 in the Mass General Brigham healthcare system. We estimated the associations of baseline characteristics with the odds of hospitalisation due to COVID-19 infection, verified by medical record review, using multivariable logistic regression. RESULTS Of 2061 patients with SARDs and COVID-19 during the Omicron era (75% female, mean age 62.2 years), 134 (6.5%) were hospitalised due to COVID-19, mostly due to respiratory symptoms (84, 63%). Of those hospitalised, 11 (8%) required mechanical ventilation and 20 (15%) died. Older age (adjusted OR (aOR) 1.05 per year), Black race (vs White race, aOR 4.15), ever smoking (vs never, aOR 1.76), CD20 inhibitor use (vs antimalarial monotherapy, aOR 2.22) and glucocorticoid use (vs non-use, aOR 2.07) were significantly associated with higher odds of hospitalisation. Female sex (vs male, aOR 0.63), booster SARS-CoV-2 vaccination (vs initial series, aOR 0.49) and vaccination within either 3 months or 3-6 months prior to infection (aOR 0.41 and aOR 0.38, respectively, vs none within 12 months) were significantly associated with lower odds of hospitalisation. CONCLUSIONS Some patients with SARDs remain at higher risk of severe COVID-19 in the Omicron era. Patients who are older, Black, have more comorbidities, use CD20 inhibitors and/or glucocorticoids, or have not been vaccinated recently may benefit from risk-mitigating strategies, including booster vaccines and pre-exposure prophylaxis.
Collapse
Affiliation(s)
- Naomi J Patel
- Harvard Medical School, Boston, MA, USA
- Rheumatology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shruthi Srivatsan
- Rheumatology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Emily N Kowalski
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Andrew King
- Rheumatology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Xiaosong Wang
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Kathleen Mm Vanni
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Grace Qian
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Jennifer S Hanberg
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Katarina J Bade
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Alene A Saavedra
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Kevin T Mueller
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Buuthien Hang
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | - Madison Negron
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jeffrey A Sparks
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Zachary S Wallace
- Harvard Medical School, Boston, MA, USA
- Rheumatology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Phillips TJ, Carlo-Stella C, Morschhauser F, Bachy E, Crump M, Trněný M, Bartlett NL, Zaucha J, Wrobel T, Offner F, Humphrey K, Relf J, Filézac de L'Etang A, Carlile DJ, Byrne B, Qayum N, Lundberg L, Dickinson M. Glofitamab in Relapsed/Refractory Mantle Cell Lymphoma: Results From a Phase I/II Study. J Clin Oncol 2025; 43:318-328. [PMID: 39365960 PMCID: PMC11771347 DOI: 10.1200/jco.23.02470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 06/25/2024] [Accepted: 08/21/2024] [Indexed: 10/06/2024] Open
Abstract
PURPOSE Patients with relapsed/refractory (R/R) mantle cell lymphoma (MCL) have a poor prognosis. The phase I/II NP30179 study (ClinicalTrials.gov identifier: NCT03075696) evaluated glofitamab monotherapy in patients with R/R B-cell lymphomas, with obinutuzumab pretreatment (Gpt) to mitigate the risk of cytokine release syndrome (CRS) with glofitamab. We present data for patients with R/R MCL. METHODS Eligible patients with R/R MCL (at least one previous therapy) received Gpt (1,000 or 2,000 mg) 7 days before the first glofitamab dose (single dose or split over 2 days if required). Glofitamab step-up dosing was administered once a day on days 8 (2.5 mg) and 15 (10 mg) of cycle 1, with a target dose of 16 or 30 mg once every 3 weeks from cycle 2 day 1 onward, for 12 cycles. Efficacy end points included investigator-assessed complete response (CR) rate, overall response rate (ORR), and duration of CR. RESULTS Of 61 enrolled patients, 60 were evaluable for safety and efficacy. Patients had received a median of two previous therapies (range, 1-5). CR rate and ORR were 78.3% (95% CI, 65.8 to 87.9) and 85.0% (95% CI, 73.4 to 92.9), respectively. In patients who had received previous treatment with a Bruton tyrosine kinase inhibitor (n = 31), CR rate was 71.0% (95% CI, 52.0 to 85.8) and ORR was 74.2% (95% CI, 55.4 to 88.1). CRS after glofitamab administration occurred in 70.0% of patients, with a lower incidence in the 2,000 mg (63.6% [grade ≥2, 22.7%]) versus 1,000 mg (87.5%; grade ≥2, 62.5%) Gpt cohort. Four adverse events led to glofitamab withdrawal (all infections). CONCLUSION Fixed-duration glofitamab induced high CR rates in heavily pretreated patients with R/R MCL; the safety profile was manageable with appropriate support.
Collapse
MESH Headings
- Humans
- Lymphoma, Mantle-Cell/drug therapy
- Lymphoma, Mantle-Cell/pathology
- Male
- Female
- Aged
- Middle Aged
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Aged, 80 and over
- Neoplasm Recurrence, Local/drug therapy
- Adult
- Cytokine Release Syndrome/prevention & control
Collapse
Affiliation(s)
- Tycel Jovelle Phillips
- University of Michigan Medical School, Ann Arbor, MI
- Current address: City of Hope National Medical Center, Duarte, CA
| | - Carmelo Carlo-Stella
- Department of Biomedical Sciences, Humanitas University and IRCCS Humanitas Research Hospital, Milano, Italy
| | | | - Emmanuel Bachy
- Hospices Civils de Lyon and Université Claude Bernard, Pierre-Bénite, France
| | - Michael Crump
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Marek Trněný
- First Faculty of Medicine, Charles University, General Hospital, Prague, Czech Republic
| | | | - Jan Zaucha
- Medical University of Gdańsk, Gdańsk, Poland
| | | | - Fritz Offner
- Department of Hematology, Universitair Ziekenhuis, Gent, Belgium
| | | | - James Relf
- Roche Products Ltd, Welwyn Garden City, United Kingdom
| | | | | | - Ben Byrne
- Roche Products Ltd, Welwyn Garden City, United Kingdom
| | - Naseer Qayum
- Roche Products Ltd, Welwyn Garden City, United Kingdom
| | | | - Michael Dickinson
- Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
5
|
Kacar M, Al-Hakim A, Savic S. Sequelae of B-Cell Depleting Therapy: An Immunologist's Perspective. BioDrugs 2025; 39:103-130. [PMID: 39680306 DOI: 10.1007/s40259-024-00696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 12/17/2024]
Abstract
B-cell depleting therapy (BCDT) has revolutionised the treatment of B-cell malignancies and autoimmune diseases by targeting specific B-cell surface antigens, receptors, ligands, and signalling pathways. This narrative review explores the mechanisms, applications, and complications of BCDT, focusing on the therapeutic advancements since the introduction of rituximab in 1997. Various monoclonal antibodies and kinase inhibitors are examined for their roles in depleting B cells through antibody-dependent and independent mechanisms. The off-target effects, such as hypogammaglobulinemia, infections, and cytokine release syndrome, are discussed, emphasising the need for immunologists to identify and help manage these complications. The increasing prevalence of BCDT has necessitated the involvement of clinical immunologists in addressing treatment-associated immunological abnormalities, including persistent hypogammaglobulinemia and neutropenia. We highlight the importance of considering underlying inborn errors of immunity (IEI) in patients presenting with these complications. Furthermore, we discuss the impact of BCDT on other immune cell populations and the challenges in predicting and managing long-term immunological sequelae. The potential for novel BCDT agents targeting the BAFF/APRIL-TACI/BCMA axis and B-cell receptor signalling pathways to treat autoimmune disorders is also explored, underscoring the rapidly evolving landscape of B-cell targeted therapies.
Collapse
Affiliation(s)
- Mark Kacar
- Department of Allergy, University Clinic Golnik, Golnik, Slovenia
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Adam Al-Hakim
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Sinisa Savic
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK.
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
- NIHR Leeds Biomedical Research Centre, Leeds, UK.
| |
Collapse
|
6
|
Abdulrasak M, Hootak S. Prolonged SARS-CoV-2 Viremia in an Immunocompromised Patient. J Med Cases 2025; 16:6-10. [PMID: 39759167 PMCID: PMC11699861 DOI: 10.14740/jmc5064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 11/09/2024] [Indexed: 01/07/2025] Open
Abstract
Immunocompromised patients, especially those receiving B-cell depleting therapies, are at risk for developing atypical presentation with regard to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, with the potential for diagnostic delay and adverse outcomes if such delay occurs. A 66-year-old female with history of granulomatosis with polyangiitis (GPA) with previous pulmonary involvement, treated with rituximab and low-dose prednisolone, presented with prolonged fever and cough after having been treated at home for a mild SARS-CoV-2 infection in early July 2023. The patient had a prolonged course over several months with constitutional symptoms such as fever, cough and malaise. During the investigation, which encompassed a wide range of microbiological and immunological tests, the patient was initially thought to have a flare of GPA which she was treated for without appreciable improvement, then for multiple microbiological organisms without appropriate resolution of the patient's symptoms. The differential diagnosis of prolonged SARS-CoV-2 infection was reconsidered in October 2023, and then confirmed by the presence of SARS-CoV-2 viremia through polymerase chain reaction (PCR) testing of the blood. The patient received a prolonged course of antiviral therapy with complete clinical, virological and radiological resolution. Prolonged SARS-CoV-2 infection with viremia in immunocompromised individuals needs to be considered on the differential diagnosis list in such patients presenting with constitutional symptoms, with PCR testing of the blood as a simple and effective way to establish the diagnosis.
Collapse
Affiliation(s)
- Mohammed Abdulrasak
- Department of Clinical Sciences, Malmo, Lund University, Malmo, Sweden
- Department of Gastroenterology and Nutrition, Skane University Hospital, Malmo, Sweden
| | - Sohail Hootak
- Department of Clinical Sciences, Malmo, Lund University, Malmo, Sweden
- Department of Gastroenterology and Nutrition, Skane University Hospital, Malmo, Sweden
| |
Collapse
|
7
|
Song H, Zhang Y, Chen Y, Zhang M, Gao F, Zhao C. Association between COVID-19 infection and uveitis flare in patients with Behcet's disease, a retrospective multicenter cohort study. Graefes Arch Clin Exp Ophthalmol 2025; 263:209-215. [PMID: 39141118 DOI: 10.1007/s00417-024-06536-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 05/18/2024] [Accepted: 05/27/2024] [Indexed: 08/15/2024] Open
Abstract
PURPOSE To explore if COVID-19 infection and its subsequent immunosuppressant adjustment as well as previous vaccination status are associated with higher risks of uveitis flare in patients with Behcet's disease. METHODS This retrospective multicenter cohort study was conducted in January 2023 among patients with Behcet's uveitis, during the second wave of the COVID-19 pandemic in China, with an anticipated sample size of 250. The primary objective was to examine the association between COVID-19 infection and the occurrence of uveitis flare. The potential impact of other exposures, including the patient's vaccination status and treatment adjustments to the risk of uveitis flare and the course of COVID-19 infection were also analyzed. RESULTS 207 patients with COVID-19 infection and 47 patients without COVID-19 infection were included. A total of 127 uveitis flares occurred in the observational period (14.29 events per 100 person-month). COVID-19 infection was found to be significantly associated with a higher rate of uveitis flare (adjusted rate ratio = 4.8, 95% CI 3.7 to 6.3, P < 0.001). However, neither systemic immunosuppressive adjustment nor COVID-19 vaccination status showed a significant association with uveitis flare or the course of COVID-19 infection. CONCLUSIONS This study provides evidence of an association between COVID-19 infection and an increased risk of uveitis flare in patients with Behcet's disease. However, there was no significant evidence to support that baseline immunosuppressive therapy regimens, treatment adjustment after COVID-19 infection, or vaccination status were associated with higher risks of uveitis flare or prolonged COVID-19 course.
Collapse
Affiliation(s)
- Hang Song
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuelun Zhang
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Chen
- Department of Ophthalmology, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, Shaanxi Province, China
| | - Meifen Zhang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fei Gao
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chan Zhao
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
8
|
Mazzotti L, Borges de Souza P, Azzali I, Angeli D, Nanni O, Sambri V, Semprini S, Bravaccini S, Cerchione C, Gaimari A, Nicolini F, Ancarani V, Martinelli G, Pasetto A, Calderon H, Juan M, Mazza M. Exploring the Relationship Between Humoral and Cellular T Cell Responses Against SARS-CoV-2 in Exposed Individuals From Emilia Romagna Region and COVID-19 Severity. HLA 2025; 105:e70011. [PMID: 39807702 PMCID: PMC11731316 DOI: 10.1111/tan.70011] [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: 07/09/2024] [Revised: 11/03/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025]
Abstract
COVID-19 remains a significant global health problem with uncertain long-term consequences for convalescents. We investigated the relationships between anti-N protein antibody levels, severe acute respiratory syndrome (SARS)-CoV-2-associated TCR repertoire parameters, HLA type and epidemiological information from three cohorts of 524 SARS-CoV-2-infected subjects subgrouped in acute phase, seronegative and seropositive convalescents from the Emilia Romagna region. Epidemiological information and anti-N antibody index were associated with TCR repertoire data. HLA type was inferred from TCR repertoire using the HLA3 tool and its association with clonal breadth (CB) and clonal depth (CD) was assessed. Age above 58 years, male and COVID-19 hospitalisation were significantly and independently associated with seropositivity (p = 0.004; p = 0.004; p = 0.04), suggesting an association between high antibody titres and symptoms' severity. As for the TCR repertoire analysis, we found no difference in CB among the cohorts, while CD was higher in seronegative than acute (p = 0.04). However, clustering analysis supported that seronegative patients are endowed with broader CB and deeper CD indicating a compensatory mechanism without effective seroconversion. The CD calculated on the TCRs associated with the single SARS-CoV-2 ORFs in convalescents is higher when compared to the acute. Lastly, we identified and reported on novel HLAs significantly associated with increased risk of hospitalisation such as HLA-C*07:02 carriers (OR = 3.9, CI = 1.1-13.4, p = 0.03) and on HLAs that associate significantly with lower or higher TCR repertoire parameters in a population exposed for the first time to SARS-CoV-2.
Collapse
Affiliation(s)
- Lucia Mazzotti
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | | | - Irene Azzali
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Davide Angeli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Oriana Nanni
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Vittorio Sambri
- Microbiology UnitThe Great Romagna Area Hub LaboratoryPievesestinaItaly
- DIMECBologna UniversityBolognaItaly
| | - Simona Semprini
- Microbiology UnitThe Great Romagna Area Hub LaboratoryPievesestinaItaly
| | - Sara Bravaccini
- Department of Medicine and SurgeryUniversity of Enna “Kore”EnnaItaly
| | - Claudio Cerchione
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Anna Gaimari
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Fabio Nicolini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Valentina Ancarani
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Giovanni Martinelli
- Department of Hematology and Sciences OncologyInstitute of Haematology “L. and A. Seràgnoli” S. Orsola, University Hospital in BolognaBolognaItaly
| | - Anna Pasetto
- Section for Cell TherapyRadiumhospitalet, Oslo University HospitalOsloNorway
- Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
| | - Hugo Calderon
- Department of ImmunologyCentre de Diagnòstic Biomèdic, Hospital Clínic of BarcelonaBarcelonaSpain
| | - Manel Juan
- Department of ImmunologyCentre de Diagnòstic Biomèdic, Hospital Clínic of BarcelonaBarcelonaSpain
| | - Massimiliano Mazza
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| |
Collapse
|
9
|
Tsukida S, Hongo M, Akasaki K, Sone T, Nishi K. COVID-19 Pneumonia Diagnosed by Bronchoalveolar Lavage Fluid, With CD4 T-cell Depletion Contributing to Prolonged Infection: Two Case Reports. Cureus 2024; 16:e74380. [PMID: 39723265 PMCID: PMC11669300 DOI: 10.7759/cureus.74380] [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] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Irrespective of the underlying disease, patients treated with cluster of differentiation 20 (CD20) antibodies have a higher risk of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) long or severe infection, and there are pitfalls in this diagnosis. We herein report two patients with COVID-19 pneumonia diagnosed by bronchoalveolar lavage fluid (BALF) during lymphoma remission. Nasopharyngeal swabs (NSs) were polymerase chain reaction (PCR)-negative for SARS-CoV-2, and the virus was only detectable in the lungs. In patients with B-cell depletion, the early performance of bronchoalveolar lavage (BAL) is important for diagnosing COVID-19 pneumonia and ruling out opportunistic infections when any evidence of suspected viral pneumonia is observed on computed tomography (CT), even if the NS specimens are PCR-negative and they have no upper respiratory symptoms. In addition, blood tests with lymphocytopenia, BALF with decreased CD4/CD8 ratio, and increased neutralizing antibody titer suggested that not only low humoral immune responses but also CD4 T-cell depletion by bendamustine were associated with virus clearance. Even if neutralizing antibodies are adequate, we must be careful of prolonged COVID-19 due to CD4 T-cell depletion and low humoral immune responses.
Collapse
Affiliation(s)
- Saya Tsukida
- Internal Medicine, Keiju Medical Center, Nanao, JPN
| | - Masato Hongo
- Internal Medicine, Fukui-ken Saiseikai Hospital, Fukui, JPN
| | - Kyota Akasaki
- Internal Medicine, Ishikawa Prefectural Central Hospital, Kanazawa, JPN
| | - Takashi Sone
- Internal Medicine, Ishikawa Prefectural Central Hospital, Kanazawa, JPN
| | - Koichi Nishi
- Internal Medicine, Ishikawa Prefectural Central Hospital, Kanazawa, JPN
| |
Collapse
|
10
|
Li YR, Lyu Z, Chen Y, Fang Y, Yang L. Frontiers in CAR-T cell therapy for autoimmune diseases. Trends Pharmacol Sci 2024; 45:839-857. [PMID: 39147651 DOI: 10.1016/j.tips.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/14/2024] [Accepted: 07/18/2024] [Indexed: 08/17/2024]
Abstract
Chimeric antigen receptor (CAR)-engineered T (CAR-T) cell therapy has demonstrated significant success in treating cancers. The potential of CAR-T cells is now being explored in the context of autoimmune diseases. Recent clinical trials have shown sustained and profound elimination of autoreactive B cells by CAR-T cells, leading to promising autoimmune disease control with minimal safety concerns. These encouraging results have inspired further investigation into CAR-T cell applications for a broader range of autoimmune diseases and the development of advanced cell products with improved efficacy and safety. In this review, we discuss the mechanisms by which CAR-T cells target autoimmune conditions, summarize current preclinical models, and highlight ongoing clinical trials, including CAR-T therapy design, clinical outcomes, and challenges. Additionally, we discuss the limitations and future directions of CAR-T therapy in the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Zibai Lyu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yuning Chen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ying Fang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
11
|
Rutskaya-Moroshan K, Abisheva S, Abisheva A, Amangeldiyeva Z, Vinnik T, Batyrkhan T. Clinical Characteristics, Prognostic Factors, and Outcomes of COVID-19 in Autoimmune Rheumatic Disease Patients: A Retrospective Case-Control Study from Astana, Kazakhstan. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1377. [PMID: 39336418 PMCID: PMC11433992 DOI: 10.3390/medicina60091377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024]
Abstract
Background: Viral infections, including coronavirus disease 2019 (COVID-19), in patients with autoimmune rheumatic diseases (AIRDs) tend to present more severe disease. This study aims to investigate the clinical characteristics and risk factors for severe infection in rheumatologic patients. Methods: We included patients with a diagnosis of AIRD and COVID-19 infection between January 2022 and July 2023. Patients with AIRDs infected with SARS-CoV-2 were matched with control patients of the general population according to age (±5 years) and sex in a 1:1 ratio. Confirmed infection was defined if a patient had a positive polymerase chain reaction (PCR) test. The severity was divided into mild, moderate, severe, and critical according to the guidelines of the United States National Institutes of Health (NIH). Results: A total of 140 individuals (37 males, 103 females; mean age 56.1 ± 11.3 years) with rheumatic disease diagnosed with COVID-19 infection were enrolled in the study. AIRDs included rheumatoid arthritis (RA) (n = 63, 45%), ankylosing spondylitis (AS) (n = 35, 25%), systemic lupus erythematosus (SLE) (n = 26, 8.6%), and systemic sclerosis (SSc) (n = 16, 11.4%). The AIRDs group had more SARS-CoV-2-related dyspnea (38.6%), arthralgia (45.7%), and depression (27.1%) than the control group (p = 0.004). The rate of lung infiltration on radiographic examination was higher in 58 (41.4%, p = 0.005) patients with rheumatic diseases than in those without them. Severe SARS-CoV-2 infection was more common in the AIRDs group than in the control group (22% vs. 12%; p = 0.043). Conclusions: Patients with AIRDs experienced more symptoms of arthralgia, depression, and dyspnea. There was a trend towards an increased severity of the disease in patients with AIRDs. Patients with arterial hypertension, diabetes, chronic lung, and kidney disease, treated with corticosteroids, had a longer duration, and high activity of autoimmune disease had an increased risk of severe COVID-19.
Collapse
Affiliation(s)
- Kristina Rutskaya-Moroshan
- Department of Family Medicine №1, NJSC «Astana Medical University», Astana 010000, Kazakhstan; (K.R.-M.); (A.A.); (Z.A.); (T.V.); (T.B.)
| | - Saule Abisheva
- Department of Family Medicine №1, NJSC «Astana Medical University», Astana 010000, Kazakhstan; (K.R.-M.); (A.A.); (Z.A.); (T.V.); (T.B.)
| | - Anilim Abisheva
- Department of Family Medicine №1, NJSC «Astana Medical University», Astana 010000, Kazakhstan; (K.R.-M.); (A.A.); (Z.A.); (T.V.); (T.B.)
| | - Zhadra Amangeldiyeva
- Department of Family Medicine №1, NJSC «Astana Medical University», Astana 010000, Kazakhstan; (K.R.-M.); (A.A.); (Z.A.); (T.V.); (T.B.)
| | - Tatyana Vinnik
- Department of Family Medicine №1, NJSC «Astana Medical University», Astana 010000, Kazakhstan; (K.R.-M.); (A.A.); (Z.A.); (T.V.); (T.B.)
- Department of Molecular Biology, Ariel University, Ariel 40700, Israel
| | - Tansholpan Batyrkhan
- Department of Family Medicine №1, NJSC «Astana Medical University», Astana 010000, Kazakhstan; (K.R.-M.); (A.A.); (Z.A.); (T.V.); (T.B.)
| |
Collapse
|
12
|
Abutiban F, Saleh K, Hayat S, Tarakmah H, Al-Herz A, Ghanem A. COVID-19 and reported mortality cases among rheumatic disease patients in Kuwait: Data from the Global Rheumatology Alliance registry. Int J Rheum Dis 2024; 27:e14771. [PMID: 37287425 DOI: 10.1111/1756-185x.14771] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/10/2023] [Accepted: 05/23/2023] [Indexed: 06/09/2023]
Abstract
Mycophenolate mofetil and rituximab have been shown to be considerably associated with poorer outcomes following SARS-CoV-2 infection. Such agents were associated with longer hospital stay as well as severe COVID-19 outcomes (infection-related complications, intensive care unit admission, and mortality). Using the data of the COVID-19 Global Rheumatology Alliance (GRA) registry of inflammatory rheumatic disease (IRD) patients in Kuwait, who had COVID-19 from March 2020 to March 2021, revealed 4 mortality cases (3 cases used CD-20 inhibitors as monotherapy and 1 case used mycophenolate mofetil/mycophenolic acid as monotherapy). This article describes the characteristics and course of disease among 4 patients with IRD who died following COVID-19 infection at Jaber Al Ahmed Hospital, Kuwait. The current series raises the intriguing prospect that IRD patients may have a varying risk of unfavorable clinical outcomes depending on the type of biological agents they were given. Rituximab and mycophenolate mofetil should be used with caution in IRD patients, particularly if they have concomitant comorbidities that put them at a high likelihood of developing severe COVID-19 outcomes.
Collapse
Affiliation(s)
- Fatemah Abutiban
- Department of Medicine, Ministry of Health, Jaber Alahmed Hospital, Kuwait City, Kuwait
| | - Khulood Saleh
- Department of Medicine, Ministry of Health, Farwaniyah Hospital, Kuwait City, Kuwait
| | - Sawsan Hayat
- Department of Medicine, Ministry of Health, Mubarak Alkabeer Hospital, Kuwait City, Kuwait
| | - Hoda Tarakmah
- Department of Medicine, Ministry of Health, Mubarak Alkabeer Hospital, Kuwait City, Kuwait
| | - Adeeba Al-Herz
- AlAmiri Hospital, Department of Medicine, Ministry of Health, Kuwait City, Kuwait
| | - Aqeel Ghanem
- Department of Medicine, Ministry of Health, Mubarak Alkabeer Hospital, Kuwait City, Kuwait
| |
Collapse
|
13
|
Najimi N, Kadi C, Elmtili N, Seghrouchni F, Bakri Y. Unravelling humoral immunity in SARS-CoV-2: Insights from infection and vaccination. Hum Antibodies 2024; 32:85-106. [PMID: 38758995 DOI: 10.3233/hab-230017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024]
Abstract
Following infection and vaccination against SARS-CoV-2, humoral components of the adaptive immune system play a key role in protecting the host. Specifically, B cells generate high-affinity antibodies against various antigens of the virus. In this review, we discuss the mechanisms of immunity initiation through both natural infection and vaccination, shedding light on the activation of B cell subsets in response to SARS-CoV-2 infection and vaccination. The innate immune system serves as the initial line of primary and nonspecific defence against viruses. However, within several days following infection or a vaccine dose, a virus-specific immune response is initiated, primarily by B cells that produce antibodies. These antibodies contribute to the resolution of the disease. Subsequently, these B cells transition into memory B cells, which play a crucial role in providing long-term immunity against the virus. CD4+ T helper cells initiate a cascade, leading to B cell somatic hypermutation, germinal center memory B cells, and the production of neutralizing antibodies. B-cell dysfunction can worsen disease severity and reduce vaccine efficacy. Notably, individuals with B cell immunodeficiency show lower IL-6 production. Furthermore, this review delves into several aspects of immune responses, such as hybrid immunity, which has shown promise in boosting broad-spectrum protection. Cross-reactive immunity is under scrutiny as well, as pre-existing antibodies can offer protection against the disease. We also decipher breakthrough infection mechanisms, especially with the novel variants of the virus. Finally, we discuss some potential therapeutic solutions regarding B cells including convalescent plasma therapy, B-1 cells, B regulatory cell (Breg) modulation, and the use of neutralizing monoclonal antibodies in combating the infection. Ongoing research is crucial to grasp population immunity trends and assess the potential need for booster doses in maintaining effective immune responses against potential viral threats.
Collapse
Affiliation(s)
- Nouhaila Najimi
- Laboratory of Human Pathologies Biology and Center of Genomic of Human Pathologies Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
- Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Chaimae Kadi
- Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Mohammed VI University of Sciences and Health, Casablanca, Morocco
- Laboratory of Biology and Health, Faculty of Sciences of Tétouan, Abdelmalek Essaâdi University, Tétouan, Morocco
| | - Noureddine Elmtili
- Laboratory of Biology and Health, Faculty of Sciences of Tétouan, Abdelmalek Essaâdi University, Tétouan, Morocco
| | - Fouad Seghrouchni
- Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Youssef Bakri
- Laboratory of Human Pathologies Biology and Center of Genomic of Human Pathologies Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| |
Collapse
|
14
|
Nguyen AA, Habiballah SB, LaBere B, Day-Lewis M, Elkins M, Al-Musa A, Chu A, Jones J, Fried AJ, McDonald D, Hoytema van Konijnenburg DP, Rockowitz S, Sliz P, Oettgen HC, Schneider LC, MacGinnitie A, Bartnikas LM, Platt CD, Ohsumi TK, Chou J. Rethinking Immunological Risk: A Retrospective Cohort Study of Severe SARS-Cov-2 Infections in Individuals With Congenital Immunodeficiencies. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:3391-3399.e3. [PMID: 37544429 PMCID: PMC10839118 DOI: 10.1016/j.jaip.2023.07.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 06/22/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Debates on the allocation of medical resources during the coronavirus disease 2019 (COVID-19) pandemic revealed the need for a better understanding of immunological risk. Studies highlighted variable clinical outcomes of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections in individuals with defects in both adaptive and innate immunity, suggesting additional contributions from other factors. Notably, none of these studies controlled for variables linked with social determinants of health. OBJECTIVE To determine the contributions of determinants of health to risk of hospitalization for SARS-CoV-2 infection among individuals with inborn errors of immunodeficiencies. METHODS This is a retrospective, single-center cohort study of 166 individuals with inborn errors of immunity, aged 2 months through 69 years, who developed SARS-CoV-2 infections from March 1, 2020, through March 31, 2022. Risks of hospitalization were assessed using a multivariable logistic regression analysis. RESULTS The risk of SARS-CoV-2-related hospitalization was associated with underrepresented racial and ethnic populations (odds ratio [OR] 4.50; 95% confidence interval [95% CI] 1.57-13.4), a diagnosis of any genetically defined immunodeficiency (OR 3.32; 95% CI 1.24-9.43), obesity (OR 4.24; 95% CI 1.38-13.3), and neurological disease (OR 4.47; 95% CI 1.44-14.3). The COVID-19 vaccination was associated with reduced hospitalization risk (OR 0.52; 95% CI 0.31-0.81). Defects in T cell and innate immune function, immune-mediated organ dysfunction, and social vulnerability were not associated with increased risk of hospitalization after controlling for covariates. CONCLUSIONS The associations between race, ethnicity, and obesity with increased risk of hospitalization for SARS-CoV-2 infection indicate the importance of variables linked with social determinants of health as immunological risk factors for individuals with inborn errors of immunity.
Collapse
Affiliation(s)
- Alan A Nguyen
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Saddiq B Habiballah
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Brenna LaBere
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Megan Day-Lewis
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Megan Elkins
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Amer Al-Musa
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Anne Chu
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Jennifer Jones
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Ari J Fried
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Douglas McDonald
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | | | - Shira Rockowitz
- Research Computing, Information Technology, Boston Children's Hospital, Boston, Mass; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Mass
| | - Piotr Sliz
- Research Computing, Information Technology, Boston Children's Hospital, Boston, Mass; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Mass; Division of Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Hans C Oettgen
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Lynda C Schneider
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Andrew MacGinnitie
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Lisa M Bartnikas
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Craig D Platt
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | | | - Janet Chou
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass.
| |
Collapse
|
15
|
Aryanian Z, Balighi K, Sajad B, Esmaeli N, Daneshpazhooh M, Mazloumi Tootoonchi N, Beigmohammadi F, Mohseni Afshar Z, Hatami P. COVID outcome in pemphigus: Does rituximab make pemphigus patients susceptible to more severe COVID-19? J Cosmet Dermatol 2023; 22:2880-2888. [PMID: 37573477 DOI: 10.1111/jocd.15958] [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: 12/24/2022] [Revised: 06/11/2023] [Accepted: 08/01/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND The COVID-19 pandemic has raised some concerns regarding the management of chronic skin diseases, especially in patients on immunosuppressive therapy including patients with pemphigus vulgaris (PV). Literature review reveals conflicting results about the effect of monoclonal antibodies such as rituximab on clinical outcome of COVID-19. OBJECTIVES To assess the reciprocal interaction of COVID-19 and pemphigus and the effect of rituximab on prognosis of COVID-19 in patients. METHODS We set up a retrospective study on adult patients with a confirmed diagnosis of pemphigus vulgaris and a history of COVID-19 with or without symptoms during 2020. RESULTS Thirty-six adults with pemphigus vulgaris and SARS-CoV-2 infection were included. The SARS-CoV-2 infection was confirmed with positive RT-PCR test results in 31 cases (86.1%) and suspected in the 5 others (13.9%). Gender, total dose of rituximab, number of rituximab cycles, and involvement of head and neck were not associated to duration of COVID-19 symptoms (p values: 0.32, 0.23, 0.84, and 0.51, respectively), severity of disease (hospitalization) (p values: 0.46, 0.39, 0.23, and 0.72, respectively), or the percentage of lung involvement on CT scan (p values: 0.07, 0.36, 0.38, and 0.09, respectively). Regarding the impact of COVID-19 on pemphigus, the majority of patients did not experience any changes in their pemphigus regarding clinical phenotype (100%) or severity (83.3%), but PV was worsened in 6 (16.9%) patients which was controlled with increasing the prednisolone dosage. CONCLUSION Rituximab appears to be safe with no increased risk of severe form of COVID-19 in patients with pemphigus vulgaris.
Collapse
Affiliation(s)
- Zeinab Aryanian
- Autoimmune Bullous Diseases Research Center, Razi hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Dermatology, Babol University of Medical Sciences, Babol, Iran
| | - Kamran Balighi
- Autoimmune Bullous Diseases Research Center, Razi hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Dermatology, School of Medicine, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Baseerat Sajad
- Autoimmune Bullous Diseases Research Center, Razi hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Esmaeli
- Autoimmune Bullous Diseases Research Center, Razi hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Dermatology, School of Medicine, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Daneshpazhooh
- Autoimmune Bullous Diseases Research Center, Razi hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Dermatology, School of Medicine, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasim Mazloumi Tootoonchi
- Autoimmune Bullous Diseases Research Center, Razi hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Dermatology, School of Medicine, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Beigmohammadi
- Autoimmune Bullous Diseases Research Center, Razi hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Mohseni Afshar
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parvaneh Hatami
- Autoimmune Bullous Diseases Research Center, Razi hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Kim D, Biancon G, Bai Z, VanOudenhove J, Liu Y, Kothari S, Gowda L, Kwan JM, Buitrago-Pocasangre NC, Lele N, Asashima H, Racke MK, Wilson JE, Givens TS, Tomayko MM, Schulz WL, Longbrake EE, Hafler DA, Halene S, Fan R. Microfluidic Immuno-Serolomic Assay Reveals Systems Level Association with COVID-19 Pathology and Vaccine Protection. SMALL METHODS 2023; 7:e2300594. [PMID: 37312418 PMCID: PMC10592458 DOI: 10.1002/smtd.202300594] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/23/2023] [Indexed: 06/15/2023]
Abstract
How to develop highly informative serology assays to evaluate the quality of immune protection against coronavirus disease-19 (COVID-19) has been a global pursuit over the past years. Here, a microfluidic high-plex immuno-serolomic assay is developed to simultaneously measure50 plasma or serum samples for50 soluble markers including 35proteins, 11 anti-spike/receptor binding domian (RBD) IgG antibodies spanningmajor variants, and controls. This assay demonstrates the quintuplicate test in a single run with high throughput, low sample volume, high reproducibilityand accuracy. It is applied to the measurement of 1012 blood samples including in-depth analysis of sera from 127 patients and 21 healthy donors over multiple time points, either with acute COVID infection or vaccination. The protein analysis reveals distinct immune mediator modules that exhibit a reduced degree of diversity in protein-protein cooperation in patients with hematologic malignancies or receiving B cell depletion therapy. Serological analysis identifies that COVID-infected patients with hematologic malignancies display impaired anti-RBD antibody response despite high level of anti-spike IgG, which can be associated with limited clonotype diversity and functional deficiency in B cells. These findings underscore the importance to individualize immunization strategies for these high-risk patients and provide an informative tool to monitor their responses at the systems level.
Collapse
Affiliation(s)
- Dongjoo Kim
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Giulia Biancon
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Zhiliang Bai
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Jennifer VanOudenhove
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Yuxin Liu
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Shalin Kothari
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Lohith Gowda
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Jennifer M Kwan
- Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | | | - Nikhil Lele
- Department of Neurology, Yale University, New Haven, CT, 06520, USA
| | | | | | | | | | - Mary M Tomayko
- Departments of Dermatology, Yale University, New Haven, CT, 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Wade L Schulz
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Erin E Longbrake
- Department of Neurology, Yale University, New Haven, CT, 06520, USA
| | - David A Hafler
- Department of Neurology, Yale University, New Haven, CT, 06520, USA
- Department of Immunobiology, Yale University, New Haven, CT, 06520, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
- Yale Center for RNA Science and Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
- Yale Cancer Center and Stem Cell Center, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
- Yale Cancer Center and Stem Cell Center, Yale School of Medicine, New Haven, CT, 06520, USA
- Human and Translational Immunology, Yale School of Medicine, New Haven, CT, 06520, USA
| |
Collapse
|
17
|
Alhumaid S, Al Mutared KM, Al Alawi Z, Sabr Z, Alkhars O, Alabdulqader M, Al Dossary N, ALShakhs FM, Majzoub RA, Alalawi YH, Al Noaim K, Alnaim AA, Al Ghamdi MA, Alahmari AA, Albattat SS, Almubarak YS, Al Abdulmohsen EM, Al Shaikh H, Alobaidan ME, Almusallam HH, Alhassan FM, Alamer MA, Al-Hajji JA, Al-Hajji DA, Alkadi AA, Al Mutair A, Rabaan AA. Severity of SARS-CoV-2 infection in children with inborn errors of immunity (primary immunodeficiencies): a systematic review. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2023; 19:69. [PMID: 37559153 PMCID: PMC10413516 DOI: 10.1186/s13223-023-00831-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 07/30/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND Inborn errors of immunity (IEIs) are considered significant challenges for children with IEIs, their families, and their medical providers. Infections are the most common complication of IEIs and children can acquire coronavirus disease 2019 (COVID-19) even when protective measures are taken. OBJECTIVES To estimate the incidence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in children with IEIs and analyse the demographic parameters, clinical characteristics and treatment outcomes in children with IEIs with COVID-19 illness. METHODS For this systematic review, we searched ProQuest, Medline, Embase, PubMed, CINAHL, Wiley online library, Scopus and Nature through the Preferred Reporting Items for Systematic Reviews and Meta Analyses (PRISMA) guideline for studies on the development of COVID-19 in children with IEIs, published from December 1, 2019 to February 28, 2023, with English language restriction. RESULTS Of the 1095 papers that were identified, 116 articles were included in the systematic review (73 case report, 38 cohort 4 case-series and 1 case-control studies). Studies involving 710 children with IEIs with confirmed COVID-19 were analyzed. Among all 710 IEIs pediatric cases who acquired SARS-CoV-2, some children were documented to be admitted to the intensive care unit (ICU) (n = 119, 16.8%), intubated and placed on mechanical ventilation (n = 87, 12.2%), suffered acute respiratory distress syndrome (n = 98, 13.8%) or died (n = 60, 8.4%). Overall, COVID-19 in children with different IEIs patents resulted in no or low severity of disease in more than 76% of all included cases (COVID-19 severity: asymptomatic = 105, mild = 351, or moderate = 88). The majority of children with IEIs received treatment for COVID-19 (n = 579, 81.5%). Multisystem inflammatory syndrome in children (MIS-C) due to COVID-19 in children with IEIs occurred in 103 (14.5%). Fatality in children with IEIs with COVID-19 was reported in any of the included IEIs categories for cellular and humoral immunodeficiencies (n = 19, 18.6%), immune dysregulatory diseases (n = 17, 17.9%), innate immunodeficiencies (n = 5, 10%), bone marrow failure (n = 1, 14.3%), complement deficiencies (n = 1, 9.1%), combined immunodeficiencies with associated or syndromic features (n = 7, 5.5%), phagocytic diseases (n = 3, 5.5%), autoinflammatory diseases (n = 2, 3%) and predominantly antibody deficiencies (n = 5, 2.5%). Mortality was COVID-19-related in a considerable number of children with IEIs (29/60, 48.3%). The highest ICU admission and fatality rates were observed in cases belonging to cellular and humoral immunodeficiencies (26.5% and 18.6%) and immune dysregulatory diseases (35.8% and 17.9%) groups, especially in children infected with SARS-CoV-2 who suffered severe combined immunodeficiency (28.6% and 23.8%), combined immunodeficiency (25% and 15%), familial hemophagocytic lymphohistiocytosis (40% and 20%), X-linked lymphoproliferative diseases-1 (75% and 75%) and X-linked lymphoproliferative diseases-2 (50% and 50%) compared to the other IEIs cases. CONCLUSION Children with IEIs infected with SARS-CoV-2 may experience higher rates of ICU admission and mortality in comparison with the immunocompetent pediatric populations. Underlying immune defects does seem to be independent risk factors for severe SARS-CoV-2 infection in children with IEIs, a number of children with SCID and CID were reported to have prolonged infections-though the number of patients is small-but especially immune dysregulation diseases (XLP1 and XLP2) and innate immunodeficiencies impairing type I interferon signalling (IFNAR1, IFNAR2 and TBK1).
Collapse
Affiliation(s)
- Saad Alhumaid
- School of Pharmacy, University of Tasmania, Hobart, 7000, Australia.
| | - Koblan M Al Mutared
- Administration of Pharmaceutical Care, Ministry of Health, 66255, Najran, Saudi Arabia
| | - Zainab Al Alawi
- Division of Allergy and Immunology, College of Medicine, King Faisal University, 31982, Hofuf, Al-Ahsa, Saudi Arabia
| | - Zainah Sabr
- Division of Allergy and Immunology, Pediatric Department, College of Medicine, King Khalid University, 62529, Abha, Saudi Arabia
| | - Ola Alkhars
- Pediatric Department, King Faisal General Hospital, Ministry of Health, 36361, Hofuf, Al-Ahsa, Saudi Arabia
| | - Muneera Alabdulqader
- Pediatric Nephrology Specialty, Pediatric Department, Medical College, King Faisal University, 31982, Hofuf, Al-Ahsa, Saudi Arabia
| | - Nourah Al Dossary
- General Surgery Department, Alomran General Hospital, Ministry of Health, 36358, Hofuf, Al-Ahsa, Saudi Arabia
| | - Fatemah M ALShakhs
- Respiratory Therapy Department, Prince Saud Bin Jalawi Hospital, Ministry of Health, 36424, Al Mubarraz, Al-Ahsa, Saudi Arabia
| | - Rabab Abbas Majzoub
- Department of Pediatrics, College of Medicine, King Faisal University, 31982, Hofuf, Al-Ahsa, Saudi Arabia
| | - Yousef Hassan Alalawi
- Ear, Nose and Throat Department, Al Jabr Hospital for Eye, Ear, Nose and Throat, Ministry of Health, 36422, Al Mubarraz, Al-Ahsa, Saudi Arabia
| | - Khalid Al Noaim
- Department of Pediatrics, College of Medicine, King Faisal University, 31982, Hofuf, Al-Ahsa, Saudi Arabia
| | - Abdulrahman A Alnaim
- Department of Pediatrics, College of Medicine, King Faisal University, 31982, Hofuf, Al-Ahsa, Saudi Arabia
| | - Mohammed A Al Ghamdi
- Department of Pediatrics, King Fahad Hospital of the University, College of Medicine, Imam Abdulrahman Bin Faisal University, 34212, Dammam, Saudi Arabia
| | - Abdulaziz A Alahmari
- Department of Pediatrics, King Fahad Hospital of the University, College of Medicine, Imam Abdulrahman Bin Faisal University, 34212, Dammam, Saudi Arabia
| | - Sawsan Sami Albattat
- College of Medicine, King Faisal University, 31982, Hofuf, Al-Ahsa, Saudi Arabia
| | - Yasin S Almubarak
- Regional Medical Supply, Al-Ahsa Health Cluster, Ministry of Health, 36361, Hofuf, Al-Ahsa, Saudi Arabia
| | | | - Hanan Al Shaikh
- Infection Prevention and Control Department, Prince Saud Bin Jalawi Hospital, Ministry of Health, 36424, Al Mubarraz, Al-Ahsa, Saudi Arabia
| | - Mortadah Essa Alobaidan
- Pharmacy Department, King Faisal General Hospital, Ministry of Health, 36361, Hofuf, Al-Ahsa, Saudi Arabia
| | - Hadi Hassan Almusallam
- Pharmacy Department, King Faisal General Hospital, Ministry of Health, 36361, Hofuf, Al-Ahsa, Saudi Arabia
| | - Fatimah Mohammed Alhassan
- Pharmacy Department, King Faisal General Hospital, Ministry of Health, 36361, Hofuf, Al-Ahsa, Saudi Arabia
| | - Mohammed Abdulhadi Alamer
- Pharmacy Department, Prince Saud Bin Jalawi Hospital, Ministry of Health, 36424, Al Mubarraz, Al-Ahsa, Saudi Arabia
| | - Jawad Ali Al-Hajji
- Primary Care Medicine, Al-Ahsa Health Cluster, Ministry of Health, 24231, Hofuf, Al-Ahsa, Saudi Arabia
| | - Duaa Ali Al-Hajji
- Nursing Department, King Faisal General Hospital, Ministry of Health, 36361, Hofuf, Al-Ahsa, Saudi Arabia
| | - Anwar Ahmed Alkadi
- Nursing Department, Prince Saud Bin Jalawi Hospital, Ministry of Health, 36424, Al Mubarraz, Al-Ahsa, Saudi Arabia
| | - Abbas Al Mutair
- Research Center, Almoosa Specialist Hospital, 36342, Al Mubarraz, Al-Ahsa, Saudi Arabia
- College of Nursing, Princess Norah Bint Abdul Rahman University, 11564, Riyadh, Saudi Arabia
- School of Nursing, University of Wollongong, Wollongong, NSW, 2522, Australia
- Nursing Department, Prince Sultan Military College of Health Sciences, 33048, Dhahran, Saudi Arabia
| | - Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, 31311, Dhahran, Saudi Arabia
- College of Medicine, Alfaisal University, 11533, Riyadh, Saudi Arabia
- Department of Public Health/Nutrition, The University of Haripur, Haripur, 22620, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
18
|
Ramasamy A, Wang C, Brode WM, Verduzco-Gutierrez M, Melamed E. Immunologic and Autoimmune-Related Sequelae of Severe Acute Respiratory Syndrome Coronavirus 2 Infection: Clinical Symptoms and Mechanisms of Disease. Phys Med Rehabil Clin N Am 2023; 34:623-642. [PMID: 37419536 PMCID: PMC10086105 DOI: 10.1016/j.pmr.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2023]
Abstract
The COVID-19 pandemic has resulted in a significant number of people developing long-term health effects of postacute sequelae SARS-CoV-2 infection (PASC). Both acute COVID-19 and PASC are now recognized as multiorgan diseases with multiple symptoms and disease causes. The development of immune dysregulation during acute COVID-19 and PASC is of high epidemiologic concern. Both conditions may also be influenced by comorbid conditions such as pulmonary dysfunction, cardiovascular disease, neuropsychiatric conditions, prior autoimmune conditions and cancer. This review discusses the clinical symptoms, pathophysiology, and risk factors that affect both acute COVID-19 and PASC.
Collapse
Affiliation(s)
- Akshara Ramasamy
- Department of Neurology, Dell Medical School, University of Texas at Austin, Health Discovery Building, 1601 Trinity Street, Austin, TX 78712, USA
| | - Chumeng Wang
- Department of Neurology, Dell Medical School, University of Texas at Austin, Health Discovery Building, 1601 Trinity Street, Austin, TX 78712, USA
| | - W Michael Brode
- Department of Internal Medicine, Dell Medical School, University of Texas at Austin, 1601 Trinity Street, Austin, TX 78712, USA
| | - Monica Verduzco-Gutierrez
- Department of Physical Medicine and Rehabilitation, University of Texas at San Antonio, 7703 Floyd Curl Drive, Mail Code 7798, San Antonio, TX 78229, USA
| | - Esther Melamed
- Department of Neurology, Dell Medical School, University of Texas at Austin, Health Discovery Building, 1601 Trinity Street, Austin, TX 78712, USA.
| |
Collapse
|
19
|
Evonuk KS, Wang S, Mattie J, Cracchiolo CJ, Mager R, Ferenčić Ž, Sprague E, Carrier B, Schofield K, Martinez E, Stewart Z, Petrosino T, Johnson GA, Yusuf I, Plaisted W, Naiman Z, Delp T, Carter L, Marušić S. Bruton's tyrosine kinase inhibition reduces disease severity in a model of secondary progressive autoimmune demyelination. Acta Neuropathol Commun 2023; 11:115. [PMID: 37438842 PMCID: PMC10337138 DOI: 10.1186/s40478-023-01614-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023] Open
Abstract
Bruton's tyrosine kinase (BTK) is an emerging target in multiple sclerosis (MS). Alongside its role in B cell receptor signaling and B cell development, BTK regulates myeloid cell activation and inflammatory responses. Here we demonstrate efficacy of BTK inhibition in a model of secondary progressive autoimmune demyelination in Biozzi mice with experimental autoimmune encephalomyelitis (EAE). We show that late in the course of disease, EAE severity could not be reduced with a potent relapse inhibitor, FTY720 (fingolimod), indicating that disease was relapse-independent. During this same phase of disease, treatment with a BTK inhibitor reduced both EAE severity and demyelination compared to vehicle treatment. Compared to vehicle treatment, late therapeutic BTK inhibition resulted in fewer spinal cord-infiltrating myeloid cells, with lower expression of CD86, pro-IL-1β, CD206, and Iba1, and higher expression of Arg1, in both tissue-resident and infiltrating myeloid cells, suggesting a less inflammatory myeloid cell milieu. These changes were accompanied by decreased spinal cord axonal damage. We show similar efficacy with two small molecule inhibitors, including a novel, highly selective, central nervous system-penetrant BTK inhibitor, GB7208. These results suggest that through lymphoid and myeloid cell regulation, BTK inhibition reduced neurodegeneration and disease progression during secondary progressive EAE.
Collapse
Affiliation(s)
| | - Sen Wang
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Josh Mattie
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - C. J. Cracchiolo
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Reine Mager
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Željko Ferenčić
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Ethan Sprague
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Brandon Carrier
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Kai Schofield
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Evelyn Martinez
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Zachary Stewart
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Tara Petrosino
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | | | - Isharat Yusuf
- Gossamer Bio, 3013 Science Park Road, Suite 200, San Diego, CA 92121 USA
| | - Warren Plaisted
- Gossamer Bio, 3013 Science Park Road, Suite 200, San Diego, CA 92121 USA
| | - Zachary Naiman
- Gossamer Bio, 3013 Science Park Road, Suite 200, San Diego, CA 92121 USA
| | - Timothy Delp
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Laura Carter
- Gossamer Bio, 3013 Science Park Road, Suite 200, San Diego, CA 92121 USA
| | - Suzana Marušić
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| |
Collapse
|
20
|
Rosenthal JA, Papa MP, Sanz M, Nicholes S, Holmberg CS, Bosque A, Keswani A, Amdur R, Lynch RM, Soriano-Sarabia N, Ein D. Antibody and T-cell responses to coronavirus disease 2019 vaccination in common variable immunodeficiency and specific antibody deficiency. Ann Allergy Asthma Immunol 2023; 130:743-751.e3. [PMID: 36736722 PMCID: PMC9891791 DOI: 10.1016/j.anai.2023.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/03/2023] [Accepted: 01/20/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND Clinical trials of the mRNA coronavirus disease 2019 (COVID-19) vaccines excluded individuals with primary antibody deficiencies. OBJECTIVE To evaluate whether antibody and T-cell responses to mRNA COVID-19 vaccination in patients with common variable immunodeficiency (CVID) and specific antibody deficiency (SAD) were comparable to those in healthy controls. METHODS We measured antibody responses against the spike glycoprotein and the receptor-binding domain (RBD) in addition to severe acute respiratory syndrome coronavirus 2 specific T-cell responses using peripheral blood mononuclear cells 2 to 8 weeks after the subjects completed the primary 2-dose vaccine series. RESULTS The study comprised 12 patients with CVID, 7 patients with SAD, and 10 controls. Individuals with CVID had lower immunoglobulin (Ig) G and Ig A levels against spike glycoprotein than did both individuals with SAD (P = .27 and P = .01, respectively) and controls (P = .01 and P = .004, respectively). The CVID group developed lower IgG titers against the RBD epitope than did the control group (P = .01). Participants with CVID had lower neutralizing titers than did the control group (P = .002). All participants with SAD developed neutralizing titers. All 3 groups (SAD, CVID, and control) developed antigen-specific CD4+ and CD8+ T-cell responses after vaccination. CONCLUSION Our results suggest that patients with CVID may have impaired antibody responses to COVID-19 vaccination but intact T-cell responses, whereas patients with SAD would be expected to have both intact antibody and T-cell responses to vaccination.
Collapse
Affiliation(s)
- Jamie A Rosenthal
- Division of Allergy-Immunology, Department of Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia.
| | - Michelle Premazzi Papa
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Marta Sanz
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Samuel Nicholes
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Carissa S Holmberg
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Alberto Bosque
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Anjeni Keswani
- Division of Allergy-Immunology, Department of Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Richard Amdur
- Feinstein Institutes for Medical Research, Northwell Health, New York, New York
| | - Rebecca M Lynch
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Natalia Soriano-Sarabia
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Daniel Ein
- Division of Allergy-Immunology, Department of Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| |
Collapse
|
21
|
Breeden M, Aitken SL, Baang JH, Gravelin M, Kaul DR, Lauring AS, Petty LA, Gregg KS. Successful Treatment of Prolonged Severe Acute Respiratory Syndrome Coronavirus 2 Infection in Patients With Immunodeficiency With Extended Nirmatrelvir/Ritonavir: Case Series. Open Forum Infect Dis 2023; 10:ofad189. [PMID: 37089775 PMCID: PMC10114524 DOI: 10.1093/ofid/ofad189] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
Immunocompromised patients with B-cell deficiencies are at risk for prolonged symptomatic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We describe 4 patients treated for B-cell malignancies with B-cell-depleting therapies who developed persistent SARS-CoV-2 infection and had resolution of symptoms following an extended course of nirmatrelvir/ritonavir.
Collapse
Affiliation(s)
- Madison Breeden
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Samuel L Aitken
- Department of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, Michigan, USA
| | - Ji Hoon Baang
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Misty Gravelin
- Michigan Institute for Clinical and Health Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel R Kaul
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Adam S Lauring
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Lindsay A Petty
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kevin S Gregg
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
22
|
Convalescent Plasma Treatment of Patients Previously Treated with B-Cell-Depleting Monoclonal Antibodies Suffering COVID-19 Is Associated with Reduced Re-Admission Rates. Viruses 2023; 15:v15030756. [PMID: 36992465 PMCID: PMC10059055 DOI: 10.3390/v15030756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/12/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023] Open
Abstract
Patients receiving treatment with B-cell-depleting monoclonal antibodies, such as anti-CD20 monoclonal antibodies, such as rituximab and obinutuzumab, either for hematological disease or another diagnosis, such as a rheumatological disease, are at an increased risk for medical complications and mortality from COVID-19. Since inconsistencies persist regarding the use of convalescent plasma (CP), especially in the vulnerable patient population that has received previous treatment with B-cell-depleting monoclonal antibodies, further studies should be performed in thisdirection. The aim of the present study was to describe the characteristics of patients with previous use of B-cell-depleting monoclonal antibodies and describe the potential beneficial effects of CP use in terms of mortality, ICU admission and disease relapse. In this retrospective cohort study, 39 patients with previous use of B-cell-depleting monoclonal antibodies hospitalized in the COVID-19 department of a tertiary hospital in Greece were recorded and evaluated. The mean age was 66.3 years and 51.3% were male. Regarding treatment for COVID-19, remdesivir was used in 89.7%, corticosteroids in 94.9% and CP in 53.8%. In-hospital mortality was 15.4%. Patients who died were more likely to need ICU admission and also had a trend towards a longer hospital stay, even though the last did not reach statistical significance. Patients treated with CP had a lower re-admission rate for COVID-19 after discharge. Further studies should be performed to identify the role of CP in patients with treatment with B-cell-depleting monoclonal antibodies suffering from COVID-19.
Collapse
|
23
|
Roppelt AA, Lebedkina MS, Chernov AA, Kruglova TS, Mukhina OA, Yukhnovskaya YD, Samedova FA, Mаrkina UA, Andrenova GV, Karaulov AV, Lysenko MA, Fomina DS. Pre-exposure prophylaxis of new COVID-19 coronavirus infection with tixagevimab/cilgavimab in adult Moscow patients with primary immunodeficiencies. TERAPEVT ARKH 2023; 95:78-84. [PMID: 37167118 DOI: 10.26442/00403660.2023.01.202088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 02/24/2023] [Indexed: 02/26/2023]
Abstract
Background. Primary immunodeficiencies (PIDs), now known as inborn errors of immunity, are a group of inherited diseases caused by defects in the genes that control the immune response. Patients with PIDs have risks of developing a severe course and/or death in COVID-19. Passive immunization with long-acting monoclonal antibodies (MABs) to SARS-CoV-2 should be considered as pre-exposure prophylaxis in patients with PIDs. Tixagevimab/cilgavimab is a combination of MABs that bind to the SARS-CoV-2 spike protein.
Aim. To evaluate the efficacy and safety of pre-exposure prophylaxis of new SARS-CoV-2 infection in PIDs with the combination of tixagevimab/cilgavimab.
Materials and methods. Forty eight patients diagnosed with PIDs were included in the study. Median follow-up after drug administration was 174 days. The total number of confirmed coronavirus infections in patients with PIDs as well as 6 months before and after administration of MAT were assessed.
Results. In the analyzed cohort, the overall incidence of COVID-19 from pandemic onset to MABs administration was 75% (36/48), with 31% (11/36) of over-infected patients having had the infection more than once. The incidence of COVID-19 immediately 6 months before the introduction of tixagevimab/cilgavimab was 40%. All patients who had COVID-19 after pre-exposure prophylaxis had a mild infection. The incidence of COVID-19 6 months after tixagevimab/cilgavimab administration significantly decreased compared to the incidence 6 months before administration (7 and 40%, respectively; p0.001).
Conclusion. The use of tixagevimab/cilgavimab in patients with PIDs is effective as pre-exposure prophylaxis and reduces the risk of severe COVID-19.
Collapse
|
24
|
Kandula UR, Tuji TS, Gudeta DB, Bulbula KL, Mohammad AA, Wari KD, Abbas A. Effectiveness of COVID-19 Convalescent Plasma (CCP) During the Pandemic Era: A Literature Review. J Blood Med 2023; 14:159-187. [PMID: 36855559 PMCID: PMC9968437 DOI: 10.2147/jbm.s397722] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 02/08/2023] [Indexed: 02/25/2023] Open
Abstract
Worldwide pandemic with coronavirus disease-2019 (COVID-19) was caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). As November 2, 2022, World Health Organization (WHO) received 628,035,553 reported incidents on COVID-19, with 6,572,800 mortalities and, with a total 12,850,970,971 vaccine doses have been delivered as of October 31, 2022. The infection can cause mild or self-limiting symptoms of pulmonary and severe infections or death may be caused by SARS-CoV-2 infection. Simultaneously, antivirals, corticosteroids, immunological treatments, antibiotics, and anticoagulants have been proposed as potential medicines to cure COVID-19 affected patients. Among these initial treatments, COVID-19 convalescent plasma (CCP), which was retrieved from COVID-19 recovered patients to be used as passive immune therapy, in which antibodies from cured patients were given to infected patients to prevent illness. Such treatment has yielded the best results in earlier with preventative or early stages of illness. Convalescent plasma (CP) is the first treatment available when infectious disease initially appears, although few randomized controlled trials (RCTs) were conducted to evaluate its effectiveness. The historical record suggests with potential benefit for other respiratory infections, as coronaviruses like Severe Acute Respiratory Syndrome-CoV-I (SARS-CoV-I) and Middle Eastern Respiratory Syndrome (MERS), though the analysis of such research is constrained by some non-randomized experiments (NREs). Rigorous studies on CP are made more demanding by the following with the immediacy of the epidemics, CP use may restrict the ability to utilize it for clinical testing, non-homogenous nature of product, highly decentralized manufacturing process; constraints with capacity to measure biologic function, ultimate availability of substitute therapies, as antivirals, purified immune globulins, or monoclonal antibodies. Though, it is still not clear how effectively CCP works among hospitalized COVID-19 patients. The current review tries to focus on its efficiency and usage in clinical scenarios and identifying existing benefits of implementation during pandemic or how it may assist with future pandemic preventions.
Collapse
Affiliation(s)
- Usha Rani Kandula
- Department of Nursing, College of Health Sciences, Arsi University, Asella, Ethiopia
| | - Techane Sisay Tuji
- Department of Nursing, College of Health Sciences, Arsi University, Asella, Ethiopia
| | | | - Kassech Leta Bulbula
- Department of Nursing, College of Health Sciences, Arsi University, Asella, Ethiopia
| | | | - Ketema Diriba Wari
- Department of Nursing, College of Health Sciences, Arsi University, Asella, Ethiopia
| | - Ahmad Abbas
- Department of Nursing, College of Health Sciences, Arsi University, Asella, Ethiopia
| |
Collapse
|
25
|
García-García A, Fortuny C, Fumadó V, Jordan I, Ruiz-López L, González-Navarro EA, Egri N, Esteve-Solé A, Luo Y, Vlagea A, Cabedo MM, Launes C, Soler A, Codina A, Juan M, Pascal M, Deyà-Martínez A, Alsina L. Acute and long-term immune responses to SARS-CoV-2 infection in unvaccinated children and young adults with inborn errors of immunity. Front Immunol 2023; 14:1084630. [PMID: 36742319 PMCID: PMC9896004 DOI: 10.3389/fimmu.2023.1084630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023] Open
Abstract
Purpose To describe SARS-CoV-2 infection outcome in unvaccinated children and young adults with inborn errors of immunity (IEI) and to compare their specific acute and long-term immune responses with a sex-, age-, and severity-matched healthy population (HC). Methods Unvaccinated IEI patients up to 22 years old infected with SARS-CoV-2 were recruited along with a cohort of HC. SARS-CoV-2 serology and ELISpot were performed in the acute phase of infection (up to 6 weeks) and at 3, 6, 9, and 12 months. Results Twenty-five IEI patients (median age 14.3 years, min.-max. range 4.5-22.8; 15/25 males; syndromic combined immunodeficiencies: 48.0%, antibody deficiencies: 16.0%) and 17 HC (median age 15.3 years, min.-max. range 5.4-20.0; 6/17 males, 35.3%) were included. Pneumonia occurred in 4/25 IEI patients. In the acute phase SARS-CoV-2 specific immunoglobulins were positive in all HC but in only half of IEI in whom it could be measured (n=17/25): IgG+ 58.8% (10/17) (p=0.009); IgM+ 41.2% (7/17)(p<0.001); IgA+ 52.9% (9/17)(p=0.003). Quantitative response (index) was also lower compared with HC: IgG IEI (3.1 ± 4.4) vs. HC (3.5 ± 1.5)(p=0.06); IgM IEI (1.9 ± 2.4) vs. HC (3.9 ± 2.4)(p=0.007); IgA IEI (3.3 ± 4.7) vs. HC (4.6 ± 2.5)(p=0.04). ELISpots positivity was qualitatively lower in IEI vs. HC (S-ELISpot IEI: 3/11, 27.3% vs. HC: 10/11, 90.9%; p=0.008; N-ELISpot IEI: 3/9, 33.3% vs. HC: 11/11, 100%; p=0.002) and also quantitatively lower (S-ELISpot IEI: mean index 3.2 ± 5.0 vs. HC 21.2 ± 17.0; p=0.001; N-ELISpot IEI: mean index 9.3 ± 16.6 vs. HC: 39.1 ± 23.7; p=0.004). As for long term response, SARS-CoV-2-IgM+ at 6 months was qualitatively lower in IEI(3/8, 37.5% vs. 9/10 HC: 90.0%; p=0.043), and quantitatively lower in all serologies IgG, M, and A (IEI n=9, 1.1 ± 0.9 vs. HC n=10, 2.1 ± 0.9, p=0.03; IEI n=9, 1.3 ± 1.5 vs. HC n=10, 2.9 ± 2.8, p=0.02; and IEI n=9, 0.6 ± 0.5 vs. HC n=10, 1.7 ± 0.8, p=0.002 -respectively) but there were no differences at remaining time points. Conclusions Our IEI pediatric cohort had a higher COVID-19 pneumonia rate than the general age-range population, with lower humoral and cellular responses in the acute phase (even lower compared to the reported IEI serological response after SARS-CoV-2 vaccination), and weaker humoral responses at 6 months after infection compared with HC.
Collapse
Affiliation(s)
- Ana García-García
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Department of Surgery and Surgical Specializations, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- Clinical Immunology Program, Hospital Sant Joan de Déu-Hospital Clínic Barcelona, Barcelona, Spain
| | - Claudia Fortuny
- Department of Surgery and Surgical Specializations, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- Paediatric Infectious Diseases Unit, Hospital Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- CIBER of Epidemiology and Public Health, Madrid, Spain
- Translational Research Network in Paediatric Infectious Diseases (RITIP), Madrid, Spain
| | - Victoria Fumadó
- Department of Surgery and Surgical Specializations, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- Paediatric Infectious Diseases Unit, Hospital Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- CIBER of Epidemiology and Public Health, Madrid, Spain
- Translational Research Network in Paediatric Infectious Diseases (RITIP), Madrid, Spain
| | - Iolanda Jordan
- Department of Surgery and Surgical Specializations, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Translational Research Network in Paediatric Infectious Diseases (RITIP), Madrid, Spain
- Paediatric Intensive Care Unit, Hospital Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
| | - Laura Ruiz-López
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Program, Hospital Sant Joan de Déu-Hospital Clínic Barcelona, Barcelona, Spain
| | | | - Natalia Egri
- Department of Immunology-CDB, Hospital Clínic-IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - Ana Esteve-Solé
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Program, Hospital Sant Joan de Déu-Hospital Clínic Barcelona, Barcelona, Spain
| | - Yiyi Luo
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Program, Hospital Sant Joan de Déu-Hospital Clínic Barcelona, Barcelona, Spain
| | - Alexandru Vlagea
- Clinical Immunology Program, Hospital Sant Joan de Déu-Hospital Clínic Barcelona, Barcelona, Spain
- Department of Immunology-CDB, Hospital Clínic-IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - Manel Monsonís Cabedo
- Department of Microbiology, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Cristian Launes
- Department of Surgery and Surgical Specializations, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- Paediatrics Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Paediatric Infectious Diseases Research Group, Institut de Recerca Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
| | - Aleix Soler
- Paediatrics Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Anna Codina
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Pathology Department and Biobank Department, Hospital Sant Joan de Deu, Esplugues de Llobregat, Barcelona, Spain
| | - Manel Juan
- Clinical Immunology Program, Hospital Sant Joan de Déu-Hospital Clínic Barcelona, Barcelona, Spain
- Department of Immunology-CDB, Hospital Clínic-IDIBAPS, Universitat de Barcelona, Barcelona, Spain
- Immunotherapy Platform, Hospital Sant Joan de Déu-Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Mariona Pascal
- Clinical Immunology Program, Hospital Sant Joan de Déu-Hospital Clínic Barcelona, Barcelona, Spain
- Department of Immunology-CDB, Hospital Clínic-IDIBAPS, Universitat de Barcelona, Barcelona, Spain
- Spanish Network for Allergy - RETIC de Asma, Reacciones Adversas y Alérgicas (ARADYAL), Madrid, Spain
| | - Angela Deyà-Martínez
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Department of Surgery and Surgical Specializations, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- Clinical Immunology Program, Hospital Sant Joan de Déu-Hospital Clínic Barcelona, Barcelona, Spain
| | - Laia Alsina
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Department of Surgery and Surgical Specializations, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- Clinical Immunology Program, Hospital Sant Joan de Déu-Hospital Clínic Barcelona, Barcelona, Spain
- Immunotherapy Platform, Hospital Sant Joan de Déu-Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
Barahona-Correa JE, Rueda-Ortiz C, López MJ, Gualtero S, Arevalo-Zambrano M. COVID-19 infection during blinatumomab therapy: Is safety a dilemma? SAGE Open Med Case Rep 2023; 11:2050313X221148548. [PMID: 36643709 PMCID: PMC9834623 DOI: 10.1177/2050313x221148548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Patients with acute lymphoblastic leukemia may be particularly vulnerable to SARS-CoV-2 infection and severe illness. The mainstay of current treatment is the use of blinatumomab in patients with refractory or relapsed B-cell precursor acute lymphoblastic leukemia. We discuss the case of a patient with relapsed acute lymphoblastic leukemia who became positive for SARS-CoV-2 during blinatumomab therapy. There are no formal recommendations on the decision to continue, withhold, or delay blinatumomab treatment in these patients. More studies exploring this issue are warranted, as SARS-CoV-2 is expected to be here to stay.
Collapse
Affiliation(s)
- Julián E Barahona-Correa
- Department of Internal Medicine,
Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogota,
Colombia
| | - Camilo Rueda-Ortiz
- Department of Internal Medicine,
Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogota,
Colombia
| | - María-José López
- Department of Internal Medicine,
Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogota,
Colombia,Division of Infectious Diseases,
Hospital Universitario San Ignacio, Bogota, Colombia
| | - Sandra Gualtero
- Department of Internal Medicine,
Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogota,
Colombia,Division of Infectious Diseases,
Hospital Universitario San Ignacio, Bogota, Colombia
| | - Mónica Arevalo-Zambrano
- Department of Internal Medicine,
Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogota,
Colombia,Department of Hematology, Hospital
Universitario San Ignacio, Bogota, Colombia,Mónica Arevalo-Zambrano, Carrera 7 #,
42-65, Bogota, Colombia.
| |
Collapse
|
27
|
Sanders H, Callas C, St. Amant H, Chung J, Dimitriades VR, Nakra NA. Case report: Clinical course and treatment of SARS-CoV-2 in a pediatric CAR-T cell recipient with persistent hypogammaglobulinemia. Front Pediatr 2023; 11:1076686. [PMID: 36969291 PMCID: PMC10036744 DOI: 10.3389/fped.2023.1076686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/21/2023] [Indexed: 03/29/2023] Open
Abstract
This report describes a pediatric patient who underwent chimeric antigen receptor (CAR) T-cell therapy for refractory B-cell acute lymphoblastic leukemia (B-ALL) four years prior, with resultant hypogammaglobulinemia for which he was receiving weekly subcutaneous immune globulin. He presented with persistent fever, dry cough, and a tingling sensation in his toes following a confirmed COVID-19 infection 3 weeks prior. His initial nasopharyngeal SARS-CoV-2 PCR was negative, leading to an extensive workup for other infections. He was ultimately diagnosed with persistent lower respiratory tract COVID-19 infection based on positive SARS-CoV-2 PCR from bronchoalveolar lavage (BAL) sampling. He was treated with a combination of remdesivir (antiviral) and casirivimab/imdevimab (combination monoclonal antibodies) with immediate improvement in fever, respiratory symptoms, and neurologic symptoms.
Collapse
Affiliation(s)
- Howard Sanders
- Department of Pediatrics, University of California Davis Medical School, Sacramento, CA, United States
| | - Christina Callas
- Department of Pediatrics, University of California Davis Medical School, Sacramento, CA, United States
| | - Helaine St. Amant
- Department of Pediatrics, University of California Davis Medical School, Sacramento, CA, United States
| | - Jong Chung
- Division of Pediatric Hematology & Oncology, Department of Pediatrics, University of California Davis Medical School, Sacramento, CA, United States
| | - Victoria R. Dimitriades
- Division of Pediatric Allergy & Immunology, Department of Pediatrics, University of California Davis Medical School, Sacramento, CA, United States
| | - Natasha A. Nakra
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of California Davis Medical School, Sacramento, CA, United States
- Correspondence: Natasha A. Nakra
| |
Collapse
|
28
|
Hussein M, Wei W, Mastey V, Sanchez RJ, Wang D, Murdock DJ, Hirshberg B, Weinreich DM, Jalbert JJ. Real-world effectiveness of casirivimab and imdevimab among patients diagnosed with COVID-19 in the ambulatory setting: a retrospective cohort study using a large claims database. BMJ Open 2022; 12:e064953. [PMID: 36535724 PMCID: PMC9764096 DOI: 10.1136/bmjopen-2022-064953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To assess the real-world effectiveness of casirivimab and imdevimab (CAS+IMD) versus no COVID-19 antibody treatment among patients diagnosed with COVID-19 in the ambulatory setting, including patients diagnosed during the Delta-dominant period prior to Omicron emergence. DESIGN Retrospective cohort study. SETTING Komodo Health closed claims database. PARTICIPANTS 13 273 128 patients diagnosed with COVID-19 (December 2020 through September 2021) were treated with CAS+IMD or untreated but treatment eligible under the Emergency Use Authorization (EUA). Each treated patient was exact and propensity score matched without replacement to up to five untreated EUA-eligible patients. INTERVENTIONS CAS+IMD. PRIMARY AND SECONDARY OUTCOME MEASURES Composite endpoint of 30-day all-cause mortality or COVID-19-related hospitalisation. Kaplan-Meier estimators were used to calculate outcome risks overall and across subgroups: age, COVID-19 vaccination status, immunocompromised status, and timing of diagnosis (December 2020 to June 2021, and July to September 2021). Cox proportional hazards models were used to estimate adjusted HRs (aHRs) and 95% CIs. RESULTS Among 75 159 CAS+IMD-treated and 1 670 338 EUA-eligible untreated patients, 73 759 treated patients were matched to 310 688 untreated patients; matched patients were ~50 years, ~60% were women and generally well balanced across risk factors. The 30-day risk of the composite outcome was 2.1% and 5.2% in the CAS+IMD-treated and CAS+IMD-untreated patients, respectively; equivalent to a 60% lower risk (aHR 0.40; 95% CI, 0.38 to 0.42). The effect of CAS+IMD was consistent across subgroups, including those who received a COVID-19 vaccine (aHR 0.48, 95% CI, 0.41 to 0.56), and those diagnosed during the Delta-dominant period (aHR 0.40, 95% CI, 0.38 to 0.42). CONCLUSIONS The real-world effectiveness of CAS+IMD is consistent with the efficacy for reducing all-cause mortality or COVID-19-related hospitalisation reported in clinical trials. Effectiveness is maintained across patient subgroups, including those prone to breakthrough infections, and was effective against susceptible variants including Delta. .
Collapse
Affiliation(s)
| | - Wenhui Wei
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Vera Mastey
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | - Degang Wang
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | | | | | | |
Collapse
|
29
|
Martínez-López D, Ferraz-Amaro I, Prieto-Peña D, Sánchez-Bilbao L, Herrero-Morant A, Álvarez-Reguera C, Benavides-Villanueva F, Corrales-Selaya C, Trigueros-Vázquez M, González-Gay MÁ, Blanco R. Coronavirus disease 2019 in patients with rheumatic immune-mediated diseases in a single University Hospital, matched case-control study and literature review. Front Med (Lausanne) 2022; 9:1056374. [PMID: 36579150 PMCID: PMC9792090 DOI: 10.3389/fmed.2022.1056374] [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: 09/28/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
Background COVID-19 may present different degrees of severity. Viral infections in patients with rheumatic inflammatory diseases (R-IMID) trend to present more severe disease. However, data comparing the severity of the disease between R-IMID and the general population are scarce. Objectives To compare predisposing factors, clinical, serological features, and severity of COVID-19 infection in patients with and without R-IMID. Methods Case-control study in a single University Hospital. We included all consecutive patients with a diagnosis of an R-IMID and COVID-19 infection up to March 31st, 2021. This cohort was compared to patients without R-IMID and not receiving immunosuppressive therapy, matched for sex and age (±5 years). Confirmed infection was defined if a patient had a positive nasopharyngeal swab for SARS-CoV-2. Severity was divided into mild, moderate, severe and critical according to the United States National Institute of Health (NIH) guidelines. Results We included 274 R-IMID patients (185 women/89 men), mean age 59.1 ± 18 years. More frequent R-IMID were: Rheumatoid arthritis (28.8%), Psoriatic Arthritis (20.1%), axial Spondyloarthritis (12.4%), Polymyalgia Rheumatica (8%) and Systemic Lupus Erythematosus (8%). Hypertension and dyslipidemia were more frequent in patients with R-IMID. Although most of the cases were mild, critical cases and deaths were more frequent in R-IMID. When adjusted by comorbidities, no statistical differences were observed. Conclusion R-IMID have a very similar clinical presentation when compared to the general population. There is a trend to an increased severity of the disease in patients with R-IMID.
Collapse
Affiliation(s)
- David Martínez-López
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Ivan Ferraz-Amaro
- Department of Rheumatology, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - Diana Prieto-Peña
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Lara Sánchez-Bilbao
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Alba Herrero-Morant
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Carmen Álvarez-Reguera
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | | | - Cristina Corrales-Selaya
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Martín Trigueros-Vázquez
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | | | - Ricardo Blanco
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain,*Correspondence: Ricardo Blanco,
| |
Collapse
|
30
|
Clonal diversity predicts persistence of SARS-CoV-2 epitope-specific T-cell response. Commun Biol 2022; 5:1351. [PMID: 36494499 PMCID: PMC9734123 DOI: 10.1038/s42003-022-04250-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 11/11/2022] [Indexed: 12/13/2022] Open
Abstract
T cells play a pivotal role in reducing disease severity during SARS-CoV-2 infection and formation of long-term immune memory. We studied 50 COVID-19 convalescent patients and found that T cell response was induced more frequently and persisted longer than circulating antibodies. We identified 756 clonotypes specific to nine CD8+ T cell epitopes. Some epitopes were recognized by highly similar public clonotypes. Receptors for other epitopes were extremely diverse, suggesting alternative modes of recognition. We tracked persistence of epitope-specific response and individual clonotypes for a median of eight months after infection. The number of recognized epitopes per patient and quantity of epitope-specific clonotypes decreased over time, but the studied epitopes were characterized by uneven decline in the number of specific T cells. Epitopes with more clonally diverse TCR repertoires induced more pronounced and durable responses. In contrast, the abundance of specific clonotypes in peripheral circulation had no influence on their persistence.
Collapse
|
31
|
Yazdanpanah N, Rezaei N. Autoimmune disorders associated with common variable immunodeficiency: prediction, diagnosis, and treatment. Expert Rev Clin Immunol 2022; 18:1265-1283. [PMID: 36197300 DOI: 10.1080/1744666x.2022.2132938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Common variable immunodeficiency (CVID) is the most common symptomatic primary immunodeficiency. Due to the wide spectrum of the CVID manifestations, the differential diagnosis becomes complicated, ends in a diagnostic delay and increased morbidity and mortality rates. Autoimmunity is one of the important complications associated with CVID. While immunoglobulin replacement therapy has considerably decreased the mortality rate in CVID patients, mainly infection-related mortality, other complications such as autoimmunity appeared prevalent and, in some cases, life threatening. AREAS COVERED In this article, genetics, responsible immune defects, autoimmune manifestations in different organs, and the diagnosis and treatment processes in CVID patients are reviewed, after searching the literature about these topics. EXPERT OPINION Considering the many phenotypes of CVID and the fact that it remained undiagnosed until older ages, it is important to include various manifestations of CVID in the differential diagnosis. Due to the different manifestations of CVID, including autoimmune diseases, interdisciplinary collaboration of physicians from different fields is highly recommended, as discussed in the manuscript. Meanwhile, it is important to determine which patients could benefit from genetic diagnostic studies since such studies are not necessary for establishing the diagnosis of CVID.
Collapse
Affiliation(s)
- Niloufar Yazdanpanah
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Jalbert JJ, Hussein M, Mastey V, Sanchez RJ, Wang D, Murdock D, Fariñas L, Bussey J, Duart C, Hirshberg B, Weinreich DM, Wei W. Effectiveness of Subcutaneous Casirivimab and Imdevimab in Ambulatory Patients with COVID-19. Infect Dis Ther 2022; 11:2125-2139. [PMID: 36181639 PMCID: PMC9526200 DOI: 10.1007/s40121-022-00691-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/22/2022] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Data on real-world effectiveness of subcutaneous (SC) casirivimab and imdevimab (CAS+IMD) for the treatment of coronavirus disease 2019 (COVID-19) are limited. The objective of this study was to assess the effectiveness of SC CAS+IMD versus no antibody treatment among patients with COVID-19. METHODS This retrospective cohort study linked Komodo Health and CDR Maguire Health and Medical data. Patients diagnosed with COVID-19 in ambulatory settings (August 1-October 30, 2021) treated with SC CAS+IMD were exact- and propensity score-matched to fewer than five untreated treatment-eligible patients and followed for the composite endpoint of 30-day all-cause mortality or COVID-19-related hospitalization. Kaplan-Meier estimators were used to calculate outcome risk overall and across subgroups. Cox proportional-hazards models were used to estimate adjusted hazard ratios (aHR) and 95% confidence intervals (CI). RESULTS Of 13,522 patients treated with CAS+IMD, 12,972 were matched to 41,848 untreated patients. The 30-day composite outcome risk was 1.9% (95% CI 1.7-2.2) and 4.4% (95% CI 4.2-4.6) in the treated and untreated cohorts, respectively; treated patients had a 49% lower relative risk of the composite outcome (aHR 0.51; 95% CI 0.46-0.58) and a 67% relative risk of 30-day mortality (aHR 0.33, 95% CI 0.18-0.60). Effectiveness was consistent across vaccination status and various subgroups. DISCUSSION Patients with COVID-19 benefitted from treatment with SC CAS+IMD versus untreated patients. The results were consistent across subgroups of patients, including older adults, immunocompromised patients, and patients vaccinated against COVID-19. Results were robust across numerous sensitivity analyses. CONCLUSION SC CAS+IMD is effective in reducing 30-day COVID-19-related hospitalization or mortality in real-world outpatient settings during the Delta-dominant period.
Collapse
Affiliation(s)
- Jessica J Jalbert
- Regeneron Pharmaceuticals, Inc., 1 Rockwood Road, Sleepy Hollow, NY, 10510, USA.
| | | | - Vera Mastey
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | - Degang Wang
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Dana Murdock
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | | | | | | | - Wenhui Wei
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| |
Collapse
|
33
|
Holroyd KB, Healy BC, Conway S, Houtchens M, Bakshi R, Bhattacharyya S, Bose G, Galetta K, Kaplan T, Severson C, Singhal T, Stazzone L, Zurawski J, Polgar-Turcsanyi M, Saxena S, Paul A, Glanz BI, Weiner HL, Chitnis T. Humoral response to COVID-19 vaccination in MS patients on disease modifying therapy: Immune profiles and clinical outcomes. Mult Scler Relat Disord 2022; 67:104079. [PMID: 35952457 PMCID: PMC9330583 DOI: 10.1016/j.msard.2022.104079] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/31/2022] [Accepted: 07/27/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Patients with multiple sclerosis (MS) on some disease modifying therapies (DMTs), particularly anti-CD20 and sphingosine-1-phosphate (S1P) modulators, are at increased risk of severe Coronavirus Disease 19 (COVID-19) and death. COVID-19 vaccinations are effective in preventing infection and severe disease, but humoral response to vaccination and outcomes of COVID-19 infection after vaccination in MS patients on DMTs remain less understood. METHODS In this retrospective single-center study, patients enrolled in the CLIMB (Comprehensive Longitudinal Investigation of Multiple Sclerosis at Brigham and Women's Hospital) study and biorepository who had been vaccinated against COVID-19 and had SARS-CoV-2 spike antibody (anti-SARS-CoV-2 S Roche-Elecsys) testing were identified and compared to healthy controls. Demographic data, serum immune profiles including lymphocyte count, B-cell count, and immunoglobulins, and clinical outcome of COVID-19 infection were collected. RESULTS 254 patients (73.2% female, mean (SD) age 52.9 (11.2) years) were identified. When controlling for age, time since vaccination, and vaccine type, patients on fingolimod, ocrelizumab, rituximab, mycophenolate mofetil, natalizumab and teriflunomide had significantly lower levels of spike antibodies compared to healthy controls (n = 34). Longer duration of treatment was associated with lower spike antibody levels in patients on anti-CD20 therapy (p = 0.016) and S1P modulators (p = 0.016) compared to healthy controls. In patients on anti-CD20 therapy, higher spike antibody levels were associated with higher CD20 cell count (p<0.001), and longer time since last anti-CD20 therapy infusion (p<0.001). 92.8% (13/14) vaccine responders (spike antibody titer >100 ug/dL) on anti-CD20 therapy demonstrated B-cell reconstitution (mean CD20 3.6%). Only 1 out of 86 patients with CD20 of 0% had a measurable spike antibody response to vaccination. During follow-up (mean 270 days), five patients were diagnosed with COVID-19 after vaccination (incidence 1.9%), all of whom had spike antibody < 20 ug/dL. No patients required ICU care or died. CONCLUSIONS Patients on some DMTs demonstrate reduced humoral immunity after Sars-CoV-2 vaccination. Longer duration of anti-CD20 therapy and reduced CD20 cell count is associated with blunted humoral response to vaccination. CD20 reconstitution >0.1% appears necessary, but not always sufficient, for humoral response to vaccination. Breakthrough COVID-19 infection in our cohort of MS patients on DMT was higher than in population studies. We propose that adjustment of B-cell therapy administration to allow for B-cell reconstitution prior to vaccination should be considered.
Collapse
Affiliation(s)
- Kathryn B Holroyd
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Brian C Healy
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Sarah Conway
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Maria Houtchens
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Rohit Bakshi
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Shamik Bhattacharyya
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Gauruv Bose
- Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Kristin Galetta
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Tamara Kaplan
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Christopher Severson
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Tarun Singhal
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Lynn Stazzone
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Jonathan Zurawski
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Mariann Polgar-Turcsanyi
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Shrishti Saxena
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Anu Paul
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Bonnie I Glanz
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Howard L Weiner
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Tanuja Chitnis
- Harvard Medical School, Boston, MA, United States; Brigham Multiple Sclerosis Center & Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States.
| |
Collapse
|
34
|
Vergidis P, Levy ER, Ristagno EH, Iyer VN, O'Horo JC, Joshi AY. COVID-19 in patients with B cell immune deficiency. J Immunol Methods 2022; 510:113351. [PMID: 36087764 PMCID: PMC9450485 DOI: 10.1016/j.jim.2022.113351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/22/2022] [Accepted: 09/02/2022] [Indexed: 12/31/2022]
Abstract
This article aims to describe the clinical manifestations and management of COVID-19 in patients with primary and secondary B cell deficient states. We describe the epidemiologic and clinical features as well as unique management paradigm including isolation precautions with COVID-19. We then focus upon primary and secondary preventive approaches including vaccination and pre- as well as post-exposure prophylaxis. Further, we elaborate upon the important disease specific risk factors in these patients and the need to conduct prospective clinical trials to develop individualized management strategies in this population.
Collapse
Affiliation(s)
- Paschalis Vergidis
- Division of Public Health, Infectious Diseases and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Emily R. Levy
- Division of Pediatric Critical Care Medicine, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA,Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth H. Ristagno
- Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Vivek N. Iyer
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - John C. O'Horo
- Division of Public Health, Infectious Diseases and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA,Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - Avni Y. Joshi
- Division of Pediatric and Adult Allergy and Immunology, Mayo Clinic, Rochester, MN, USA,Corresponding author at: Mayo Clinic Childrens Center, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
35
|
Göschl L, Mrak D, Grabmeier-Pfistershammer K, Stiasny K, Haslacher H, Schneider L, Deimel T, Kartnig F, Tobudic S, Aletaha D, Burgmann H, Bonelli M, Pickl WF, Förster-Waldl E, Scheinecker C, Vossen MG. Reactogenicity and immunogenicity of the second COVID-19 vaccination in patients with inborn errors of immunity or mannan-binding lectin deficiency. Front Immunol 2022; 13:974987. [PMID: 36189225 PMCID: PMC9515892 DOI: 10.3389/fimmu.2022.974987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/16/2022] [Indexed: 11/29/2022] Open
Abstract
Background Patients with inborn errors of immunity (IEI) are at increased risk for severe courses of SARS-CoV-2 infection. COVID-19 vaccination provides effective protection in healthy individuals. However, it remains unclear whether vaccination is efficient and safe in patients with constitutional dysfunctions of the immune system. Thus, we analyzed the humoral response, adverse reactions and assessed the disease activity of the underlying disease after COVID-19 vaccination in a cohort of patients suffering from IEIs or mannan-binding lectin deficiency (MBLdef). Methods Vaccination response was assessed after basic immunization using the Elecsys anti-SARS-CoV-2 S immunoassay and via Vero E6 cell based assay to detect neutralization capabilities. Phenotyping of lymphocytes was performed by flow cytometry. Patient charts were reviewed for disease activity, autoimmune phenomena as well as immunization status and reactogenicity of the vaccination. Activity of the underlying disease was assessed using a patient global numeric rating scale (NRS). Results Our cohort included 11 individuals with common variable immunodeficiency (CVID), one patient with warts hypogammaglobulinemia immunodeficiency myelokathexis (WHIM) syndrome, two patients with X-linked agammaglobulinemia (XLA), one patient with Muckle Wells syndrome, two patients with cryopyrin-associated periodic syndrome, one patient with Interferon-gamma (IFN-gamma) receptor defect, one patient with selective deficiency in pneumococcal antibody response combined with a low MBL level and seven patients with severe MBL deficiency. COVID-19 vaccination was generally well tolerated with little to no triggering of autoimmune phenomena. 20 out of 26 patients developed an adequate humoral vaccine response. 9 out of 11 patients developed a T cell response comparable to healthy control subjects. Tested immunoglobulin replacement therapy (IgRT) preparations contained Anti-SARS-CoV-2 S antibodies implicating additional protection through IgRT. Summary In summary the data support the efficacy and safety of a COVID-19 vaccination in patients with IEIs/MBLdef. We recommend evaluation of the humoral immune response and testing for virus neutralization after vaccination in this cohort.
Collapse
Affiliation(s)
- Lisa Göschl
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Daniel Mrak
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | | | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Lisa Schneider
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Thomas Deimel
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Felix Kartnig
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Selma Tobudic
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Heinz Burgmann
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Michael Bonelli
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Winfried F. Pickl
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Förster-Waldl
- Division of Neonatology, Pediatric Intensive Care and Neuropediatrics with Centre for Congenital Immunodeficiencies & Jeffrey Modell Center Vienna, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Clemens Scheinecker
- Division of Rheumatology, University Clinics of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Matthias Gerhard Vossen
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- *Correspondence: Matthias Gerhard Vossen,
| |
Collapse
|
36
|
Kim D, Biancon G, Bai Z, VanOudenhove J, Liu Y, Kothari S, Gowda L, Kwan JM, Buitrago-Pocasangre NC, Lele N, Asashima H, Racke MK, Wilson JE, Givens TS, Tomayko MM, Schulz WL, Longbrake EE, Hafler DA, Halene S, Fan R. Microfluidic immuno-serology assay revealed a limited diversity of protection against COVID-19 in patients with altered immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.08.31.506117. [PMID: 36093346 PMCID: PMC9460970 DOI: 10.1101/2022.08.31.506117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The immune response to SARS-CoV-2 for patients with altered immunity such as hematologic malignancies and autoimmune disease may differ substantially from that in general population. These patients remain at high risk despite wide-spread adoption of vaccination. It is critical to examine the differences at the systems level between the general population and the patients with altered immunity in terms of immunologic and serological responses to COVID-19 infection and vaccination. Here, we developed a novel microfluidic chip for high-plex immuno-serological assay to simultaneously measure up to 50 plasma or serum samples for up to 50 soluble markers including 35 plasma proteins, 11 anti-spike/RBD IgG antibodies spanning all major variants, and controls. Our assay demonstrated the quintuplicate test in a single run with high throughput, low sample volume input, high reproducibility and high accuracy. It was applied to the measurement of 1,012 blood samples including in-depth analysis of sera from 127 patients and 21 healthy donors over multiple time points, either with acute COVID infection or vaccination. The protein association matrix analysis revealed distinct immune mediator protein modules that exhibited a reduced degree of diversity in protein-protein cooperation in patients with hematologic malignancies and patients with autoimmune disorders receiving B cell depletion therapy. Serological analysis identified that COVID infected patients with hematologic malignancies display impaired anti-RBD antibody response despite high level of anti-spike IgG, which could be associated with limited clonotype diversity and functional deficiency in B cells and was further confirmed by single-cell BCR and transcriptome sequencing. These findings underscore the importance to individualize immunization strategy for these high-risk patients and provide an informative tool to monitor their responses at the systems level.
Collapse
Affiliation(s)
- Dongjoo Kim
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Giulia Biancon
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Zhiliang Bai
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Jennifer VanOudenhove
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yuxin Liu
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Shalin Kothari
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Lohith Gowda
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Jennifer M Kwan
- Cardiovascular Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | | | - Nikhil Lele
- Department of Neurology, Yale University, New Haven, CT 06520, USA
| | | | | | | | | | - Mary M Tomayko
- Departments of Dermatology, Yale University, New Haven, CT 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Wade L Schulz
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Erin E Longbrake
- Department of Neurology, Yale University, New Haven, CT 06520, USA
| | - David A Hafler
- Department of Neurology, Yale University, New Haven, CT 06520, USA
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Center for RNA Science and Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
- Human and Translational Immunology, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
37
|
Calabrese C, Kirchner E, Villa-Forte A, Hajj-Ali RA, Moss BP, Fernandez JP, Calabrese L. Early experience with tixagevimab/cilgavimab pre-exposure prophylaxis in patients with immune-mediated inflammatory disease undergoing B cell depleting therapy and those with inborn errors of humoral immunity. RMD Open 2022; 8:rmdopen-2022-002557. [PMID: 36123015 PMCID: PMC9485640 DOI: 10.1136/rmdopen-2022-002557] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2022] [Indexed: 11/03/2022] Open
Affiliation(s)
- Cassandra Calabrese
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, Ohio, USA
| | - Elizabeth Kirchner
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, Ohio, USA
| | - Alexandra Villa-Forte
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, Ohio, USA
| | - Rula A Hajj-Ali
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, Ohio, USA
| | - Brandon P Moss
- Mellen Center for Multiple Sclerosis Treatment and Research, Cleveland Clinic, Cleveland, Ohio, USA
| | - James P Fernandez
- Department of Allergy and Clinical Immunology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Leonard Calabrese
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
38
|
McCulloch L, Mouat IC, South K, McColl BW, Allan SM, Smith CJ. Stroke-induced changes to immune function and their relevance to increased risk of severe COVID-19 disease. DISCOVERY IMMUNOLOGY 2022; 1:kyac004. [PMID: 38566903 PMCID: PMC10917238 DOI: 10.1093/discim/kyac004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 07/07/2022] [Accepted: 08/01/2022] [Indexed: 04/04/2024]
Abstract
As the COVID-19 pandemic moves towards endemic disease, it remains of key importance to identify groups of individuals vulnerable to severe infection and understand the biological factors that mediate this risk. Stroke patients are at increased risk of developing severe COVID-19, likely due to stroke-induced alterations to systemic immune function. Furthermore, immune responses associated with severe COVID-19 in patients without a history of stroke parallel many of the immune alterations induced by stroke, possibly resulting in a compounding effect that contributes to worsened disease severity. In this review, we discuss the changes to systemic immune function that likely contribute to augmented COVID-19 severity in patients with a history of stroke and the effects of COVID-19 on the immune system that may exacerbate these effects.
Collapse
Affiliation(s)
- Laura McCulloch
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Isobel C Mouat
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Kieron South
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Barry W McColl
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Stuart M Allan
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Craig J Smith
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Greater Manchester Comprehensive Stroke Centre, Manchester Centre for Clinical Neurosciences, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, Salford, UK
| |
Collapse
|
39
|
Stefanski AL, Rincon-Arevalo H, Schrezenmeier E, Karberg K, Szelinski F, Ritter J, Chen Y, Meisel C, Jahrsdörfer B, Ludwig C, Schrezenmeier H, Lino AC, Dörner T. Persistent but atypical germinal center reaction among 3rd SARS-CoV-2 vaccination after rituximab exposure. Front Immunol 2022; 13:943476. [PMID: 36032111 PMCID: PMC9399943 DOI: 10.3389/fimmu.2022.943476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Durable vaccine-mediated immunity relies on the generation of long-lived plasma cells and memory B cells (MBCs), differentiating upon germinal center (GC) reactions. SARS-CoV-2 mRNA vaccination induces a strong GC response in healthy volunteers (HC), but limited data is available about response longevity upon rituximab treatment. Methods We evaluated humoral and cellular responses upon 3rd vaccination in seven patients with rheumatoid arthritis (RA) who initially mounted anti-spike SARS-CoV-2 IgG antibodies after primary 2x vaccination and got re-exposed to rituximab (RTX) 1-2 months after the second vaccination. Ten patients with RA on other therapies and ten HC represented the control groups. As control for known long-lived induced immunity, we analyzed humoral and cellular tetanus toxoid (TT) immune responses in steady-state. Results After 3rd vaccination, 5/7 seroconverted RTX patients revealed lower anti-SARS-CoV-2 IgG levels but similar neutralizing capacity compared with HC. Antibody levels after 3rd vaccination correlated with values after 2nd vaccination. Despite significant reduction of circulating total and antigen-specific B cells in RTX re-exposed patients, we observed the induction of IgG+ MBCs upon 3rd vaccination. Notably, only RTX treated patients revealed a high amount of IgA+ MBCs before and IgA+ plasmablasts after 3rd vaccination. IgA+ B cells were not part of the steady state TT+ B cell pool. TNF-secretion and generation of effector memory CD4 spike-specific T cells were significantly boosted upon 3rd vaccination. Summary On the basis of pre-existing affinity matured MBCs within primary immunisation, RTX re-exposed patients revealed a persistent but atypical GC immune response accompanied by boosted spike-specific memory CD4 T cells upon SARS-CoV-2 recall vaccination.
Collapse
Affiliation(s)
- Ana-Luisa Stefanski
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
- *Correspondence: Ana-Luisa Stefanski,
| | - Hector Rincon-Arevalo
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Instituto de Investigaciones Médicas, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Eva Schrezenmeier
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health Charité Universitätsmedizin Berlin, Berlin Institute of Health (BIH) Academy, Berlin, Germany
| | - Kirsten Karberg
- Rheumatology Outpatient Office RheumaPraxis Steglitz, Berlin, Germany
| | - Franziska Szelinski
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
| | - Jacob Ritter
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health Charité Universitätsmedizin Berlin, Berlin Institute of Health (BIH) Academy, Berlin, Germany
| | - Yidan Chen
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
| | - Christian Meisel
- Department of Medical Immunology, Charité University Medicine and Labor Berlin-Charité Vivantes, Berlin, Germany
| | - Bernd Jahrsdörfer
- Institute of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Carolin Ludwig
- Institute of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Hubert Schrezenmeier
- Institute of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | | | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
| |
Collapse
|
40
|
Markarian NM, Galli G, Patel D, Hemmings M, Nagpal P, Berghuis AM, Abrahamyan L, Vidal SM. Identifying Markers of Emerging SARS-CoV-2 Variants in Patients With Secondary Immunodeficiency. Front Microbiol 2022; 13:933983. [PMID: 35847101 PMCID: PMC9283111 DOI: 10.3389/fmicb.2022.933983] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/31/2022] [Indexed: 12/03/2022] Open
Abstract
Since the end of 2019, the world has been challenged by the coronavirus disease 2019 (COVID-19) pandemic. With COVID-19 cases rising globally, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to evolve, resulting in the emergence of variants of interest (VOI) and of concern (VOC). Of the hundreds of millions infected, immunodeficient patients are one of the vulnerable cohorts that are most susceptible to this virus. These individuals include those with preexisting health conditions and/or those undergoing immunosuppressive treatment (secondary immunodeficiency). In these cases, several researchers have reported chronic infections in the presence of anti-COVID-19 treatments that may potentially lead to the evolution of the virus within the host. Such variations occurred in a variety of viral proteins, including key structural ones involved in pathogenesis such as spike proteins. Tracking and comparing such mutations with those arisen in the general population may provide information about functional sites within the SARS-CoV-2 genome. In this study, we reviewed the current literature regarding the specific features of SARS-CoV-2 evolution in immunocompromised patients and identified recurrent de novo amino acid changes in virus isolates of these patients that can potentially play an important role in SARS-CoV-2 pathogenesis and evolution.
Collapse
Affiliation(s)
- Nathan M. Markarian
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill University Research Centre on Complex Traits, Montréal, QC, Canada
- Swine and Poultry Infectious Diseases Research Center and Research Group on Infectious Diseases in Production Animals, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Gaël Galli
- McGill University Research Centre on Complex Traits, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- CNRS, ImmunoConcEpT, UMR 5164, Université de Bordeaux, Bordeaux, France
- CHU de Bordeaux, FHU ACRONIM, Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares Est/Sud-Ouest, Bordeaux, France
| | - Dhanesh Patel
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill University Research Centre on Complex Traits, Montréal, QC, Canada
| | - Mark Hemmings
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| | - Priya Nagpal
- Department of Pharmacology, McGill University, Montréal, QC, Canada
| | | | - Levon Abrahamyan
- Swine and Poultry Infectious Diseases Research Center and Research Group on Infectious Diseases in Production Animals, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Silvia M. Vidal
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill University Research Centre on Complex Traits, Montréal, QC, Canada
| |
Collapse
|
41
|
Farooq MM, Miloslavsky EM, Konikov N, Ahmed AR. Use of rituximab in the treatment of mucous membrane pemphigoid: An analytic review. Autoimmun Rev 2022; 21:103119. [PMID: 35688385 DOI: 10.1016/j.autrev.2022.103119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/15/2022] [Indexed: 11/26/2022]
Abstract
Mucous Membrane Pemphigoid (MMP) is a potentially fatal mucocutaneous autoimmune blistering disease. Autoantibodies are produced against various components of the dermo-epidermal or mucosal-submucosal junction are referred to as basement membrane zone (BMZ). The hallmark is deposition of of Ig and C3 on the perilesional tissues and in some patients detection of anti-BMZ autoantibodies. A unique characteristic of MMP is that as the blisters or erosions heal, they leave irreversible scarring. This scarring results in serious and catastrophic sequelae that affect the quality of life. Conventional therapy consists of anti-inflammatory and immunosuppressive agents (ISA). In patients who fail conventional therapy or develop significant side effects to them, rituximab (RTX) has been used off label. In this review, the clinical outcomes of patients with MMP treated with RTX were studied. 124 patients were identified, 47.58% being male. 72 patients were treated by the Lymphoma Protocol and 51 by Rheumatoid Arthritis (RA) protocol. Follow up for the entire cohort was 36 months (range 0.5-72). On follow-up 64 patients (51.61%) achieved complete clinical remission (CR) off therapy, 25 patients (20.16%) were in CR on therapy, 5 patients (4.03%) were non-responders, and 9 patients (7.25%) were failures. 52 patients (41.93%) experienced a relapse, after 36 months follow-up. Duration between last RTX infusion and relapse was 10.5 months (range 1-30). Most patients with relapses were treated with additional RTX. A statistically significant better outcome was observed in patients treated with RTX as monotherapy compared to those who received RTX with ISA. Clinical outcomes in patients treated with Lymphoma protocol were better than RA protocol at a statistically significant level. Data on CD20+ B cell depletion and repopulation was limited. Interestingly relapses were seen in patients with CD20+ B cell depletion and after repopulation. In the final analysis, 89 patients (71.77%) were in complete remission. Data in this review indicated that RTX was a useful agent to treat MMP. While a randomized control trial may not be practically possible, better and disease specific protocols need to be developed. When publishing, authors should attempt to provide complete and detailed information. In doing so, they will benefit their colleagues and the patients with MMP they treat with RTX.
Collapse
Affiliation(s)
| | - Eli M Miloslavsky
- Massachusetts General Hospital, Department of Medicine, Division of Rheumatology, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02215, USA
| | - Nellie Konikov
- Boston VA health Care System, Jamaica Plain, Boston, MA 02130, USA
| | - A Razzaque Ahmed
- Center for Blistering Diseases, Boston, MA 02135, USA; Department of Dermatology, Tufts University School of Medicine, Boston, MA 02111, USA.
| |
Collapse
|
42
|
Dioverti MV, Gaston DC, Morris CP, Huff CA, Jain T, Jones R, Anders V, Lederman H, Saunders J, Mostafa HH, Avery RK. Combination Therapy With Casirivimab/Imdevimab and Remdesivir for Protracted SARS-CoV-2 Infection in B-cell-Depleted Patients. Open Forum Infect Dis 2022; 9:ofac064. [PMID: 35663288 PMCID: PMC9154336 DOI: 10.1093/ofid/ofac064] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/04/2022] [Indexed: 12/12/2022] Open
Abstract
Profoundly B-cell-depleted patients can have prolonged severe acute respiratory syndrome coronavirus 2 infections with evidence of active viral replication, due to inability to mount an adequate humoral response to clear the virus. We present 3 B-cell-depleted patients with prolonged coronavirus disease 2019 infection who were successfully treated with a combination of casirivimab/imdevimab and remdesivir.
Collapse
Affiliation(s)
- M Veronica Dioverti
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - David C Gaston
- Division of Medical Microbiology, Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - C Paul Morris
- Division of Medical Microbiology, Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| | - Carol Ann Huff
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Tania Jain
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Richard Jones
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Viki Anders
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Howard Lederman
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Division of Pediatric Allergy, Immunology and Rheumatology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jacqueline Saunders
- Oncology Investigational Drug Service Pharmacy, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Heba H Mostafa
- Division of Medical Microbiology, Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Robin K Avery
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
43
|
Stefanski A, Rincon‐Arevalo H, Schrezenmeier E, Karberg K, Szelinski F, Ritter J, Jahrsdörfer B, Schrezenmeier H, Ludwig C, Sattler A, Kotsch K, Chen Y, Claußnitzer A, Haibel H, Proft F, Guerra G, Durek P, Heinrich F, Ferreira‐Gomes M, Burmester GR, Radbruch A, Mashreghi M, Lino AC, Dörner T. B Cell Numbers Predict Humoral and Cellular Response Upon SARS-CoV-2 Vaccination Among Patients Treated With Rituximab. Arthritis Rheumatol 2022; 74:934-947. [PMID: 34962360 PMCID: PMC9011692 DOI: 10.1002/art.42060] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/25/2021] [Accepted: 12/22/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Patients with autoimmune inflammatory rheumatic diseases receiving rituximab (RTX) therapy are at higher risk of poor COVID-19 outcomes and show substantially impaired humoral immune response to anti-SARS-CoV-2 vaccine. However, the complex relationship between antigen-specific B cells and T cells and the level of B cell repopulation necessary to achieve anti-vaccine responses remain largely unknown. METHODS Antibody responses to SARS-CoV-2 vaccines and induction of antigen-specific B and CD4/CD8 T cell subsets were studied in 19 patients with rheumatoid arthritis (RA) or antineutrophil cytoplasmic antibody-associated vasculitis receiving RTX, 12 patients with RA receiving other therapies, and 30 healthy controls after SARS-CoV-2 vaccination with either messenger RNA or vector-based vaccines. RESULTS A minimum of 10 B cells per microliter (0.4% of lymphocytes) in the peripheral circulation appeared to be required for RTX-treated patients to mount seroconversion to anti-S1 IgG upon SARS-CoV-2 vaccination. RTX-treated patients who lacked IgG seroconversion showed reduced receptor-binding domain-positive B cells (P = 0.0005), a lower frequency of Tfh-like cells (P = 0.0481), as well as fewer activated CD4 (P = 0.0036) and CD8 T cells (P = 0.0308) compared to RTX-treated patients who achieved IgG seroconversion. Functionally relevant B cell depletion resulted in impaired interferon-γ secretion by spike-specific CD4 T cells (P = 0.0112, r = 0.5342). In contrast, antigen-specific CD8 T cells were reduced in both RA patients and RTX-treated patients, independently of IgG formation. CONCLUSION In RTX-treated patients, a minimum of 10 B cells per microliter in the peripheral circulation is a candidate biomarker for a high likelihood of an appropriate cellular and humoral response after SARS-CoV-2 vaccination. Mechanistically, the data emphasize the crucial role of costimulatory B cell functions for the proper induction of CD4 responses propagating vaccine-specific B cell and plasma cell differentiation.
Collapse
Affiliation(s)
- Ana‐Luisa Stefanski
- Charité Universitätsmedizin Berlin and Deutsches RheumaforschungszentrumBerlinGermany
| | - Hector Rincon‐Arevalo
- Charité Universitätsmedizin Berlin and Deutsches Rheumaforschungszentrum, Berlin, Germany, and Universidad de AntioquiaMedellínColombia
| | - Eva Schrezenmeier
- Charité Universitätsmedizin, Berlin, Deutsches Rheumaforschungszentrum Berlin and Berlin Institute of Health BIH AcademyBerlinGermany
| | | | - Franziska Szelinski
- Charité Universitätsmedizin Berlin and Deutsches RheumaforschungszentrumBerlinGermany
| | - Jacob Ritter
- Charité Universitätsmedizin Berlin and Berlin Institute of Health BIH AcademyBerlinGermany
| | - Bernd Jahrsdörfer
- Ulm University, Ulm, Germany, Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden‐Württemberg–Hessen, and University Hospital UlmUlmGermany
| | - Hubert Schrezenmeier
- Ulm University, Ulm, Germany, Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden‐Württemberg–Hessen, and University Hospital UlmUlmGermany
| | - Carolin Ludwig
- Ulm University, Ulm, Germany, Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden‐Württemberg–Hessen, and University Hospital UlmUlmGermany
| | | | | | - Yidan Chen
- Charité Universitätsmedizin Berlin and Deutsches RheumaforschungszentrumBerlinGermany
| | | | | | | | | | - Pawel Durek
- Deutsches RheumaforschungszentrumBerlinGermany
| | | | | | - Gerd R. Burmester
- Charité Universitätsmedizin Berlin and Deutsches RheumaforschungszentrumBerlinGermany
| | | | | | | | - Thomas Dörner
- Charité Universitätsmedizin Berlin and Deutsches RheumaforschungszentrumBerlinGermany
| |
Collapse
|
44
|
Manzano GS, Rice DR, Klawiter EC, Matiello M, Gillani RL, Tauhid SS, Bakshi R, Mateen FJ. Anti-SARS-CoV-2 monoclonal antibodies for the treatment of active COVID-19 in multiple sclerosis: An observational study. Mult Scler 2022; 28:1146-1150. [PMID: 35475382 DOI: 10.1177/13524585221092309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Monoclonal antibodies (mAbs) against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) received emergency use authorization for the acute treatment of COVID-19. We are not aware of published data on their use in immunosuppressed people with multiple sclerosis (pwMS). We report 23 pwMS (mean age = 49 years, ocrelizumab (n = 19), fingolimod (n = 2), vaccinated with at least an initial series (n = 19)) who received mAb for acute COVID-19. Following mAb receipt, approximately half recovered in <7 days (48%). There were no adverse events or deaths. Use of mAb for pwMS treated with fingolimod or ocrelizumab was not observed to be harmful and is likely helpful for treatment of acute COVID-19.
Collapse
Affiliation(s)
- Giovanna S Manzano
- Division of Neuroimmunology and Neuroinfectious Diseases, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA/Harvard Medical School, Boston, MA, USA
| | - Dylan R Rice
- Division of Neuroimmunology and Neuroinfectious Diseases, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Eric C Klawiter
- Division of Neuroimmunology and Neuroinfectious Diseases, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA/Harvard Medical School, Boston, MA, USA
| | - Marcelo Matiello
- Division of Neuroimmunology and Neuroinfectious Diseases, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA/Harvard Medical School, Boston, MA, USA
| | - Rebecca L Gillani
- Division of Neuroimmunology and Neuroinfectious Diseases, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA/Harvard Medical School, Boston, MA, USA
| | - Shahamat S Tauhid
- Harvard Medical School, Boston, MA, USA/Brigham Multiple Sclerosis Center, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Rohit Bakshi
- Harvard Medical School, Boston, MA, USA/Brigham Multiple Sclerosis Center, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Farrah J Mateen
- Division of Neuroimmunology and Neuroinfectious Diseases, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA/Harvard Medical School, Boston, MA, USA
| |
Collapse
|
45
|
Stefanski AL, Rincon-Arevalo H, Schrezenmeier E, Karberg K, Szelinski F, Ritter J, Chen Y, Jahrsdörfer B, Ludwig C, Schrezenmeier H, Lino AC, Dörner T. B Cell Characteristics at Baseline Predict Vaccination Response in RTX Treated Patients. Front Immunol 2022; 13:822885. [PMID: 35514962 PMCID: PMC9063458 DOI: 10.3389/fimmu.2022.822885] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/28/2022] [Indexed: 12/14/2022] Open
Abstract
Background Vaccination is considered as most efficient strategy in controlling SARS-CoV-2 pandemic spread. Nevertheless, patients with autoimmune inflammatory rheumatic diseases receiving rituximab (RTX) are at increased risk to fail humoral and cellular responses upon vaccination. The ability to predict vaccination responses is essential to guide adequate safety and optimal protection in these patients. Methods B- and T- cell data before vaccination were evaluated for characteristics predicting vaccine responses in altogether 15 patients with autoimmune inflammatory rheumatic diseases receiving RTX. Eleven patients with rheumatoid arthritis (RA) on other therapies, 11 kidney transplant recipients (KTR) on regular immunosuppression and 15 healthy controls (HC) served as controls. A multidimensional analysis of B cell subsets via UMAP algorithm and a correlation matrix were performed in order to identify predictive markers of response in patients under RTX therapy. Results Significant differences regarding absolute B cell counts and specific subset distribution pattern between the groups were identified at baseline. In this context, the majority of B cells from vaccination responders of the RTX group (RTX IgG+) were naïve and transitional B cells, whereas vaccination non-responders (RTX IgG-) carried preferentially plasmablasts and double negative (CD27-IgD-) B cells. Moreover, there was a positive correlation between neutralizing antibodies and B cells expressing HLA-DR and CXCR5 as well as an inverse correlation with CD95 expression and CD21low expression by B cells among vaccination responders. Summary Substantial repopulation of the naïve B cell compartment after RTX therapy appeared to be essential for an adequate vaccination response, which seem to require the additional capability of antigen presentation and germinal center formation. Moreover, expression of exhaustion markers represent negative predictors of vaccination responses.
Collapse
Affiliation(s)
- Ana-Luisa Stefanski
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
| | - Hector Rincon-Arevalo
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Instituto de Investigaciones Médicas, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Eva Schrezenmeier
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health Charité Universitätsmedizin Berlin, Berlin Institute of Health (BIH) Academy, Berlin, Germany
| | - Kirsten Karberg
- Rheumatology Outpatient Office RheumaPraxis Steglitz Berlin, Berlin, Germany
| | - Franziska Szelinski
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
| | - Jacob Ritter
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health Charité Universitätsmedizin Berlin, Berlin Institute of Health (BIH) Academy, Berlin, Germany
| | - Yidan Chen
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
| | - Bernd Jahrsdörfer
- Institute of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg–Hessen and University Hospital Ulm, Ulm, Germany
| | - Carolin Ludwig
- Institute of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg–Hessen and University Hospital Ulm, Ulm, Germany
| | - Hubert Schrezenmeier
- Institute of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg–Hessen and University Hospital Ulm, Ulm, Germany
| | | | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
| |
Collapse
|
46
|
Scarpa R, Dell'Edera A, Felice C, Buso R, Muscianisi F, Finco Gambier R, Toffolo S, Grossi U, Giobbia M, Barberio G, Landini N, Facchini C, Agostini C, Rattazzi M, Cinetto F. Impact of Hypogammaglobulinemia on the Course of COVID-19 in a Non-Intensive Care Setting: A Single-Center Retrospective Cohort Study. Front Immunol 2022; 13:842643. [PMID: 35359947 PMCID: PMC8960988 DOI: 10.3389/fimmu.2022.842643] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/01/2022] [Indexed: 12/11/2022] Open
Abstract
Background Severity and mortality of COVID-19 largely depends on the ability of the immune system to clear the virus. Among various comorbidities potentially impacting on this process, the weight and the consequences of an antibody deficiency have not yet been clarified. Methods We used serum protein electrophoresis to screen for hypogammaglobulinemia in a cohort of consecutive adult patients with COVID-19 pneumonia, hospitalized in non-intensive care setting between December 2020 and January 2021. The disease severity, measured by a validated score and by the need for semi intensive (sICU) or intensive care unit (ICU) admission, and the 30-day mortality was compared between patients presenting hypogammaglobulinemia (HYPO) and without hypogammaglobulinemia (no-HYPO). Demographics, comorbidities, COVID-19 specific treatment during the hospital stay, disease duration, complications and laboratory parameters were also evaluated in both groups. Results We enrolled 374 patients, of which 39 represented the HYPO cohort (10.4%). In 10/39 the condition was previously neglected, while in the other 29/39 hematologic malignancies were common (61.5%); 2/39 were on regular immunoglobulin replacement therapy (IgRT). Patients belonging to the HYPO group more frequently developed a severe COVID-19 and more often required sICU/ICU admission than no-HYPO patients. IgRT were administered in 8/39 during hospitalization; none of them died or needed sICU/ICU. Among HYPO cohort, we observed a significantly higher prevalence of neoplastic affections, of active oncologic treatment and bronchiectasis, together with higher prevalence of viral and bacterial superinfections, mechanical ventilation, convalescent plasma and SARS-CoV-2 monoclonal antibodies administration during hospital stay, and longer disease duration. Multivariate logistic regression analysis and Cox proportional hazard regression confirmed the impact of hypogammaglobulinemia on the COVID-19 severity and the probability of sICU/ICU admission. The analysis of the mortality rate in the whole cohort showed no significant difference between HYPO and no-HYPO. Conclusions Hypogammaglobulinemia, regardless of its cause, in COVID-19 patients hospitalized in a non-intensive care setting was associated to a more severe disease course and more frequent admission to s-ICU/ICU, particularly in absence of IgRT. Our findings emphasize the add-value of routine serum protein electrophoresis evaluation in patients admitted with COVID-19 to support clinicians in patient care and to consider IgRT initiation during hospitalization.
Collapse
Affiliation(s)
- Riccardo Scarpa
- Internal Medicine I, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy.,Department of Medicine, University of Padova, Padua, Italy
| | - Alessandro Dell'Edera
- Internal Medicine I, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy.,Department of Medicine, University of Padova, Padua, Italy
| | - Carla Felice
- Internal Medicine I, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy.,Department of Medicine, University of Padova, Padua, Italy
| | - Roberta Buso
- Internal Medicine I, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy
| | - Francesco Muscianisi
- Internal Medicine I, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy.,Department of Medicine, University of Padova, Padua, Italy
| | - Renato Finco Gambier
- Internal Medicine I, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy.,Department of Medicine, University of Padova, Padua, Italy
| | - Sara Toffolo
- Internal Medicine I, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy.,Department of Medicine, University of Padova, Padua, Italy
| | - Ugo Grossi
- Department of Surgery, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy
| | - Mario Giobbia
- Infectious Diseases Unit, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy
| | - Giuseppina Barberio
- Laboratory Medicine, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy
| | - Nicholas Landini
- Radiology Unit, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy
| | - Cesarina Facchini
- Internal Medicine I, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy
| | - Carlo Agostini
- Internal Medicine I, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy.,Department of Medicine, University of Padova, Padua, Italy
| | - Marcello Rattazzi
- Internal Medicine I, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy.,Department of Medicine, University of Padova, Padua, Italy
| | - Francesco Cinetto
- Internal Medicine I, Ca' Foncello Hospital, Azienda Unità Locale Socio Sanitaria n. 2 (AULSS2) Marca Trevigiana, Treviso, Italy.,Department of Medicine, University of Padova, Padua, Italy
| |
Collapse
|
47
|
Grainger R, Kim AHJ, Conway R, Yazdany J, Robinson PC. COVID-19 in people with rheumatic diseases: risks, outcomes, treatment considerations. Nat Rev Rheumatol 2022; 18:191-204. [PMID: 35217850 PMCID: PMC8874732 DOI: 10.1038/s41584-022-00755-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2022] [Indexed: 02/06/2023]
Abstract
The COVID-19 pandemic has brought challenges for people with rheumatic disease in addition to those faced by the general population, including concerns about higher risks of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and poor outcomes of COVID-19. The data that are now available suggest that rheumatic disease is associated with a small additional risk of SARS-CoV-2 infection, and that outcomes of COVID-19 are primarily influenced by comorbidities and particular disease states or treatments. Despite considerable advances in our knowledge of which therapeutic agents provide benefits in COVID-19, and of what constitutes effective vaccination strategies, the specific considerations that apply to people with rheumatic disease are yet to be definitively addressed. An overview of the most important COVID-19 studies to date that relate to people with rheumatic disease can contribute to our understanding of the clinical-care requirements of this population.
Collapse
Affiliation(s)
- Rebecca Grainger
- Department of Medicine, University of Otago, Wellington, New Zealand
| | - Alfred H J Kim
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Richard Conway
- Department of Rheumatology, St James's Hospital, Dublin, Ireland
| | - Jinoos Yazdany
- Division of Rheumatology, Department of Medicine, San Francisco General Hospital, University of California, San Francisco, CA, USA
| | - Philip C Robinson
- University of Queensland School of Clinical Medicine, Faculty of Medicine, Herston, Queensland, Australia.
- Royal Brisbane & Women's Hospital, Metro North Hospital & Health Service, Herston Road, Herston, Queensland, Australia.
| |
Collapse
|
48
|
Muacevic A, Adler JR, Tyker A, Bauer Ventura I, Lee C, Jablonski R, Vij R, Chung J, Strek M, Adegunsoye A. Mortality Risk From COVID-19 Among Unvaccinated Subjects With Autoimmune Phenotypes of Interstitial Lung Disease. Cureus 2022; 14:e23808. [PMID: 35530871 PMCID: PMC9067351 DOI: 10.7759/cureus.23808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND The impact of the severe acute respiratory syndrome-associated coronavirus 2 (SARS-CoV-2) virus on patients with interstitial lung disease (ILD) remains poorly understood. As patients with ILD often have severe underlying lung parenchymal involvement, and immunosuppressive therapy is common in this population, they are presumed to be at high risk for severe coronavirus disease 2019 (COVID-19) pneumonitis. Our aim was to explore demographic and clinical differences between those with ILD who tested positive for the SARS-CoV-2 virus compared to those with ILD who did not. METHODS In this retrospective cohort study, we identified adult, unvaccinated patients evaluated at the University of Chicago in 2020 who were enrolled in the ILD registry, and stratified by SARS-CoV-2 seropositive status. We then compared baseline clinical characteristics between SARS-CoV-2 seropositive and SARS-CoV-2 seronegative patients and assessed immunosuppressive therapy that the patient may have been on since ILD diagnosis. C-reactive protein and leukocyte subsets were evaluated at COVID diagnosis compared to the time of baseline ILD evaluation as were pulmonary function testing. Variable comparisons were determined by two-sided t-tests or chi-square tests as appropriate, and logistic regression models were fitted to assess the odds of death from COVID-19 using generalized linear models with maximum-likelihood estimation. RESULTS Of the 309 individuals with ILD in our cohort, 6.8% (n=21) tested positive for SARS-CoV-2. Those who were SARS-CoV-2 positive were younger (57 years vs 66 years; P=0.002), had baseline higher total lung capacity (81% vs 73%, P=0.045), similar forced vital capacity (71% vs. 67%, P=0.37), and similar diffusion capacity of carbon monoxide (71% vs. 62%, P=0.10) at baseline. Among patients with ILD and COVID-19, 67% had received immunosuppressive therapies compared to 74% of those with ILD without COVID-19. Those with ILD and COVID-19 were also more likely to have had a diagnosis of autoimmune-related ILD (connective tissue disease-ILD or interstitial pneumonia with autoimmune features) (62% vs 38%, P=0.029). Overall, the mortality hazard was highest among unvaccinated subjects with autoimmune-related ILD who had COVID-19 (OR=9.6, 95% CI=1.7-54.0; P=0.01). DISCUSSION SARS-CoV-2 is prevalent in ILD, and may put unvaccinated adults who are younger, with autoimmune ILD, and on immunosuppressive therapy at higher risk. This suggests a need for COVID-19 vaccinations and therapy (inpatient and outpatient) for this group of patients at high risk for COVID-19. Larger studies are needed to fully explore the relationship between ILD and immunosuppressive therapy in COVID-19.
Collapse
|
49
|
Patel NJ, Stone JH. Expert Perspective: Management of Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Arthritis Rheumatol 2022; 74:1305-1317. [PMID: 35289109 DOI: 10.1002/art.42114] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/11/2022] [Accepted: 03/09/2022] [Indexed: 12/30/2022]
Abstract
The antineutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV) comprise a major subset of diseases that cause destructive inflammation of small and medium-sized blood vessels. Although these conditions have a predilection for pulmonary and renal involvement, they are in fact protean diseases that can involve essentially any organ system. AAV is among the most difficult rheumatic diseases to diagnose and treat. Therapy for AAV has evolved over the past two decades. Rituximab, an anti-CD20 monoclonal antibody, is now the preferred agent for remission induction in conjunction with a reduced-dose glucocorticoid taper. Rituximab is also often a key therapy for remission maintenance. Glucocorticoid toxicity reduction has become a major priority for treatment regimens. Avacopan, an important new adjunct to remission induction therapy, may reduce glucocorticoid use and its resulting toxicity. The role of avacopan as a remission maintenance agent requires further study. The duration of immunosuppression following remission is guided by a number of factors, including the patient's overall clinical state, the degree of damage from previous disease activity, the tolerability of remission maintenance medications, and SARS-CoV-2 vaccination and immunity status. Certain features, including history of previous relapse, the presence of ANCA directed against proteinase 3, and a diagnosis of granulomatosis with polyangiitis, favor prolonged remission maintenance therapy. The interval between rituximab doses can usually be lengthened over time during the maintenance phase.
Collapse
|
50
|
Waldman M, Soler MJ, García-Carro C, Lightstone L, Turner-Stokes T, Griffith M, Torras J, Martinez Valenzuela L, Bestard O, Geddes C, Flossmann O, Budge KL, Cantarelli C, Fiaccadori E, Delsante M, Morales E, Gutierrez E, Niño-Cruz JA, Martinez-Rueda AJ, Comai G, Bini C, La Manna G, Slon MF, Manrique J, Avello A, Fernandez-Prado R, Ortiz A, Marinaki S, Martin Varas CR, Rabasco Ruiz C, Sierra-Carpio M, García-Agudo R, Fernández Juárez G, Hamilton AJ, Bruchfeld A, Chrysochou C, Howard L, Sinha S, Leach T, Agraz Pamplona I, Maggiore U, Cravedi P. COVID-19 in Patients with Glomerular Disease: Follow-Up Results from the IRoc-GN International Registry. KIDNEY360 2022; 3:293-306. [PMID: 35373130 PMCID: PMC8967646 DOI: 10.34067/kid.0006612021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/03/2021] [Indexed: 02/08/2023]
Abstract
Background The acute and long-term effects of severe acute respiratory syndrome coronavirus 2 infection in individuals with GN are still unclear. To address this relevant issue, we created the International Registry of COVID-19 infection in GN. Methods We collected serial information on kidney-related and -unrelated outcomes from 125 GN patients (63 hospitalized and 62 outpatients) and 83 non-GN hospitalized patients with coronavirus disease 2019 (COVID-19) and a median follow-up period of 6.4 (interquartile range 2.3-9.6) months after diagnosis. We used logistic regression for the analyses of clinical outcomes and linear mixed models for the longitudinal analyses of eGFR. All multiple regression models were adjusted for age, sex, ethnicity, and renin-angiotensin-aldosterone system inhibitor use. Results After adjustment for pre-COVID-19 eGFR and other confounders, mortality and AKI did not differ between GN patients and controls (adjusted odds ratio for AKI=1.28; 95% confidence interval [CI], 0.46 to 3.60; P=0.64). The main predictor of AKI was pre-COVID-19 eGFR (adjusted odds ratio per 1 SD unit decrease in eGFR=3.04; 95% CI, 1.76 to 5.28; P<0.001). GN patients developing AKI were less likely to recover pre-COVID-19 eGFR compared with controls (adjusted 6-month post-COVID-19 eGFR=0.41; 95% CI, 0.25 to 0.56; times pre-COVID-19 eGFR). Shorter duration of GN diagnosis, higher pre-COVID-19 proteinuria, and diagnosis of focal segmental glomerulosclerosis or minimal change disease were associated with a lower post-COVID-19 eGFR. Conclusions Pre-COVID-19 eGFR is the main risk factor for AKI regardless of GN diagnosis. However, GN patients are at higher risk of impaired eGFR recovery after COVID-19-associated AKI. These patients (especially those with high baseline proteinuria or a diagnosis of focal segmental glomerulosclerosis or minimal change disease) should be closely monitored not only during the acute phases of COVID-19 but also after its resolution.
Collapse
Affiliation(s)
- Meryl Waldman
- Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Maria Jose Soler
- Servei Nefrologia, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Grup de Recerca de Nefrología, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Clara García-Carro
- Servei Nefrologia, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Grup de Recerca de Nefrología, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Liz Lightstone
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, United Kingdom
- Imperial College Healthcare NHS Trust Renal and Transplant Centre, Hammersmith Hospital, London, United Kingdom
| | - Tabitha Turner-Stokes
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, United Kingdom
- Imperial College Healthcare NHS Trust Renal and Transplant Centre, Hammersmith Hospital, London, United Kingdom
| | - Megan Griffith
- Imperial College Healthcare NHS Trust Renal and Transplant Centre, Hammersmith Hospital, London, United Kingdom
| | - Joan Torras
- Nephrology Department, Bellvitge University Hospital, Clinical Science Department, Barcelona University, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Laura Martinez Valenzuela
- Nephrology Department, Bellvitge University Hospital, Clinical Science Department, Barcelona University, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Oriol Bestard
- Servei Nefrologia, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Grup de Recerca de Nefrología, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Colin Geddes
- Glasgow Renal and Transplant Unit, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Oliver Flossmann
- Department of Nephrology, Royal Berkshire Hospital, Reading, United Kingdom
| | - Kelly L. Budge
- Department of Medicine, Renal Division, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Chiara Cantarelli
- Dipartimento di Medicina e Chirurgia, Università di Parma, UO Nefrologia, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Enrico Fiaccadori
- Dipartimento di Medicina e Chirurgia, Università di Parma, UO Nefrologia, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Marco Delsante
- Dipartimento di Medicina e Chirurgia, Università di Parma, UO Nefrologia, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Enrique Morales
- Departamento de Nefrología, Hospital Universitario 12 de Octubre/Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Eduardo Gutierrez
- Departamento de Nefrología, Hospital Universitario 12 de Octubre/Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Jose A. Niño-Cruz
- Departamento de Nefrología y Metabolismo Mineral Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Armando J. Martinez-Rueda
- Departamento de Nefrología y Metabolismo Mineral Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Giorgia Comai
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria de Bologna, Alma Mater Studiorum University of Bologna, Italy, Bologna, Italy
| | - Claudia Bini
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria de Bologna, Alma Mater Studiorum University of Bologna, Italy, Bologna, Italy
| | - Gaetano La Manna
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria de Bologna, Alma Mater Studiorum University of Bologna, Italy, Bologna, Italy
| | | | | | - Alejandro Avello
- Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, Madrid, Spain
- Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Raul Fernandez-Prado
- Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, Madrid, Spain
- Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Alberto Ortiz
- Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, Madrid, Spain
- Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Smaragdi Marinaki
- Clinic of Nephrology and Renal Transplantation, NKUA, Medical School, Laiko General Hospital, Athens, Greece
| | | | | | | | - Rebeca García-Agudo
- Nephrology Department La Mancha-Centro Hospital, Alcázar de San Juan, Ciudad Real, Spain
| | | | | | - Annette Bruchfeld
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Department of Renal Medicine, Karolinska University Hospital and CLINTEC Karolinska Institutet, Stockholm, Sweden
| | - Constantina Chrysochou
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, United Kingdom
| | - Lilian Howard
- Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Smeeta Sinha
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, United Kingdom
| | - Tim Leach
- Portsmouth Hospitals NHS Trust, Portsmouth, United Kingdom
| | - Irene Agraz Pamplona
- Servei Nefrologia, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Grup de Recerca de Nefrología, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Umberto Maggiore
- Dipartimento di Medicina e Chirurgia, Università di Parma, UO Nefrologia, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Paolo Cravedi
- Department of Medicine, Renal Division, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|