1
|
Chong EA, Kumashie KG, Chong ER, Fabrizio J, Gupta A, Svoboda J, Barta SK, Walsh KM, Napier EB, Lundberg RK, Nasta SD, Gerson JN, Landsburg DJ, Gonzalez J, Gaano A, Weirick ME, McAllister CM, Awofolaju M, John GN, Kammerman SC, Novacek J, Pajarillo R, Lundgreen KA, Tanenbaum N, Gouma S, Drapeau EM, Adamski S, D’Andrea K, Pattekar A, Hicks A, Korte S, Sharma H, Herring S, Williams JC, Hamilton JT, Bates P, Hensley SE, Prak ETL, Greenplate AR, Wherry EJ, Schuster SJ, Ruella M, Vella LA. Immunologic Predictors of Vaccine Responsiveness in Patients With Lymphoma and Chronic Lymphocytic Leukemia. J Infect Dis 2024; 230:15-27. [PMID: 39052709 PMCID: PMC11272091 DOI: 10.1093/infdis/jiae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/13/2024] [Accepted: 02/24/2024] [Indexed: 03/06/2024] Open
Abstract
Patients with B-cell lymphomas have altered cellular components of vaccine responses due to malignancy and therapy, and the optimal timing of vaccination relative to therapy remains unknown. Severe acute respiratory syndrome coronavirus 2 vaccines created an opportunity for new insights in vaccine timing because patients were challenged with a novel antigen across multiple phases of treatment. We studied serologic messenger RNA vaccine response in retrospective and prospective cohorts with lymphoma and chronic lymphocytic leukemia, paired with clinical and research immune parameters. Reduced serologic response was observed more frequently during active treatment, but nonresponse was also common within observation and posttreatment groups. Total immunoglobulin A and immunoglobulin M correlated with successful vaccine response. In individuals treated with anti-CD19-directed chimeric antigen receptor-modified T cells, nonresponse was associated with reduced B and T follicular helper cells. Predictors of vaccine response varied by disease and therapeutic group, and therefore further studies of immune health during and after cancer therapies are needed to individualize vaccine timing.
Collapse
Affiliation(s)
- Elise A Chong
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | | | - Emeline R Chong
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
| | - Joseph Fabrizio
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
| | - Aditi Gupta
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | - Jakub Svoboda
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | - Stefan K Barta
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | - Kristy M Walsh
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
| | - Ellen B Napier
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
| | - Rachel K Lundberg
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
| | - Sunita D Nasta
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | - James N Gerson
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | - Daniel J Landsburg
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | | | | | | | | | | | - Gavin N John
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia
| | - Shane C Kammerman
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia
| | - Josef Novacek
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia
| | | | | | | | | | | | - Sharon Adamski
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | - Kurt D’Andrea
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | - Ajinkya Pattekar
- Center for Cellular Immunotherapies
- Department of Pathology and Laboratory Medicine
| | - Amanda Hicks
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | - Scott Korte
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | - Harsh Sharma
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | - Sarah Herring
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | | | - Jacob T Hamilton
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
| | | | | | | | | | - E John Wherry
- Institute for Immunology
- Department of Pathology and Laboratory Medicine
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Stephen J Schuster
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
| | - Marco Ruella
- The Richard Berman Family Innovations Center in CLL and Lymphomas, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania
- Center for Cellular Immunotherapies
- Institute for Immunology
| | - Laura A Vella
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia
- Department of Pathology and Laboratory Medicine
| |
Collapse
|
2
|
Baxter RM, Cabrera-Martinez B, Ghosh T, Rester C, Moreno MG, Borko TL, Selva S, Fleischer CL, Haakonsen N, Mayher A, Bowhay E, Evans C, Miller TM, Huey L, McWilliams J, van Bokhoven A, Deane KD, Knight V, Jordan KR, Ghosh D, Klarquist J, Kedl RM, Piquet AL, Hsieh EWY. SARS-CoV-2 Vaccine-Elicited Immunity after B Cell Depletion in Multiple Sclerosis. Immunohorizons 2024; 8:254-268. [PMID: 38483384 PMCID: PMC10985059 DOI: 10.4049/immunohorizons.2300108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/15/2024] [Indexed: 04/04/2024] Open
Abstract
The impact of B cell deficiency on the humoral and cellular responses to SARS-CoV2 mRNA vaccination remains a challenging and significant clinical management question. We evaluated vaccine-elicited serological and cellular responses in 1) healthy individuals who were pre-exposed to SARS-CoV-2 (n = 21), 2) healthy individuals who received a homologous booster (mRNA, n = 19; or Novavax, n = 19), and 3) persons with multiple sclerosis on B cell depletion therapy (MS-αCD20) receiving mRNA homologous boosting (n = 36). Pre-exposure increased humoral and CD4 T cellular responses in immunocompetent individuals. Novavax homologous boosting induced a significantly more robust serological response than mRNA boosting. MS-α CD20 had an intact IgA mucosal response and an enhanced CD8 T cell response to mRNA boosting compared with immunocompetent individuals. This enhanced cellular response was characterized by the expansion of only effector, not memory, T cells. The enhancement of CD8 T cells in the setting of B cell depletion suggests a regulatory mechanism between B and CD8 T cell vaccine responses.
Collapse
Affiliation(s)
- Ryan M. Baxter
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | | | - Tusharkanti Ghosh
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO
| | - Cody Rester
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Miguel Guerrero Moreno
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Tyler L. Borko
- Department of Neurology, Sections of Neuroimmunology, Neuroinfectious Disease, and Neurohospitalist, University of Colorado School of Medicine, Aurora, CO
| | - Sean Selva
- Department of Neurology, Sections of Neuroimmunology, Neuroinfectious Disease, and Neurohospitalist, University of Colorado School of Medicine, Aurora, CO
| | - Chelsie L. Fleischer
- Department of Medicine, Division of Rheumatology, University of Colorado, School of Medicine, Aurora, CO
| | - Nicola Haakonsen
- Department of Medicine, Division of Infectious Diseases, University of Colorado, School of Medicine, Aurora, CO
| | - Ariana Mayher
- Allergy and Immunology Research, Research Institute, Children’s Hospital Colorado, Aurora, CO
| | - Emily Bowhay
- Allergy and Immunology Research, Research Institute, Children’s Hospital Colorado, Aurora, CO
| | - Courtney Evans
- Allergy and Immunology Research, Research Institute, Children’s Hospital Colorado, Aurora, CO
| | - Todd M. Miller
- Analytics Resource Center, Children’s Hospital Colorado, Aurora, CO
| | - Leah Huey
- Department of Pediatrics, Section of Allergy and Immunology, University of Colorado, School of Medicine, Aurora, CO
| | - Jennifer McWilliams
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Adrie van Bokhoven
- Department of Pathology, Section of Pathology Shared Resource, University of Colorado, Aurora, CO
| | - Kevin D. Deane
- Department of Medicine, Division of Rheumatology, University of Colorado, School of Medicine, Aurora, CO
| | - Vijaya Knight
- Department of Pediatrics, Section of Allergy and Immunology, University of Colorado, School of Medicine, Aurora, CO
| | - Kimberly R. Jordan
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Debashis Ghosh
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO
| | - Jared Klarquist
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Ross M. Kedl
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Amanda L. Piquet
- Department of Neurology, Sections of Neuroimmunology, Neuroinfectious Disease, and Neurohospitalist, University of Colorado School of Medicine, Aurora, CO
| | - Elena W. Y. Hsieh
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
- Department of Pediatrics, Section of Allergy and Immunology, University of Colorado, School of Medicine, Aurora, CO
| |
Collapse
|
3
|
Rivera-Izquierdo M, Morales-Portillo A, Guerrero-Fernández de Alba I, Fernández-Martínez NF, Schoenenberger-Arnaiz JA, Barranco-Quintana JL, Valero-Ubierna C. Vaccination strategies for patients under monoclonal antibody and other biological treatments: an updated comprehensive review based on EMA authorisations to January 2024. Expert Rev Vaccines 2024; 23:887-910. [PMID: 39258843 DOI: 10.1080/14760584.2024.2401839] [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: 05/08/2024] [Revised: 08/13/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024]
Abstract
INTRODUCTION Monoclonal antibodies (mAbs) and other biological agents are being increasingly approved in the last years with very different indications. Their highly heterogeneous immunosuppressive effects, mechanisms of action and pharmacokinetics require comprehensive individualized vaccination schedules. AREAS COVERED Vaccination for immunocompromised patients. Prevention and treatment with mAbs and other biological therapies. EXPERT OPINION Current recommendations on vaccine schedules for patients under mAbs or other biological treatments are based on expert opinions and are not individualized according to each vaccine and treatment. No studies are focusing on the high heterogeneity of these agents, which are exponentially developed and used for many different indications. Recent paradigm changes in vaccine development (boosted by the COVID-19 pandemic) and in the mAbs use for prophylactic purposes (changing 'vaccination' by 'immunization' schedules) has been witnessed in the last years. We aimed at collecting all mAbs used for treatment or prevention, approved as of 1 January 2024, by the EMA. Based on available data on mAbs and vaccines, we propose a comprehensive guide for personalizing vaccination. Recent vaccine developments and current population strategies (e.g. zoster vaccination or prophylactic nirsevimab) are discussed. This review aims to be a practical guideline for professionals working in vaccine consultations for immunosuppressed patients.
Collapse
Affiliation(s)
- Mario Rivera-Izquierdo
- Service of Preventive Medicine and Public Health, Hospital Universitario San Cecilio, Granada, Spain
- Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain
- Instituto de investigación biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Arturo Morales-Portillo
- Service of Pharmacy, Hospital Universitari Arnau de Vilanova, Lleida, Spain
- Pharmacoepidemiology and Pharmacodynamics Group, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | | | - Nicolás Francisco Fernández-Martínez
- Instituto de investigación biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Andalusian School of Public Health (EASP), Granada, Spain
| | - Joan Antoni Schoenenberger-Arnaiz
- Service of Pharmacy, Hospital Universitari Arnau de Vilanova, Lleida, Spain
- Pharmacoepidemiology and Pharmacodynamics Group, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - José Luis Barranco-Quintana
- Service of Preventive Medicine and Public Health, Hospital Universitario Reina Sofía, Córdoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC) Córdoba, Spain
- Expert Committee on Andalusian Vaccine Plan, Consejería de Salud y Familias, Junta de Andalucía, Sevilla, Spain
| | - Carmen Valero-Ubierna
- Service of Preventive Medicine and Public Health, Hospital Universitario San Cecilio, Granada, Spain
| |
Collapse
|
4
|
Welte T, Westermann L, Kappes J, Schramm MA, Bemtgen X, Staudacher DL, Hug MJ, Venhoff N, Arnold F. Identification of Covariates Modulating B-Cell Repopulation Kinetics in Subjects Receiving Rituximab Treatment. Arthritis Rheumatol 2023; 75:2045-2053. [PMID: 37276446 DOI: 10.1002/art.42625] [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: 12/29/2022] [Revised: 05/15/2023] [Accepted: 05/30/2023] [Indexed: 06/07/2023]
Abstract
OBJECTIVE B-cell depletion using the anti-CD20 monoclonal antibody rituximab is a cornerstone in the therapeutic concept of multiple autoimmune diseases. B-cell depletion is associated with a higher risk for severe infections, and the time span of B-cell repopulation differs greatly between individuals. Data on factors influencing B-cell repopulation kinetics are limited. This study aims to identify patient-specific and therapy-associated covariates that modulate B-cell repopulation. METHODS This single-center retrospective observational study presents data of 839 subjects receiving 2,017 courses of rituximab for autoimmune diseases. Assessed covariates are patient-specific factors (sex, age, kidney function, and underlying disease) and co-immunosuppression with common agents (azathioprine, cyclosporine A, cyclophosphamide, hydroxychloroquine, methotrexate, mycophenolate mofetil, tacrolimus, and corticosteroids). The primary end point is the time to B-cell repopulation (≥5/μl). The secondary end point is the time to B-cell reconstitution (≥50/μl). Multivariate time-to-event analysis and logistic regression models were applied to estimate the influence of covariates. RESULTS Age over 60 years (hazard ratio [HR] 0.71 for repopulation, P = 0.008), impaired kidney function (HR 0.72, P = 0.001), antineutrophil cytoplasmic antibody-associated vasculitis (HR 0.61, P < 0.001), solid organ transplantation (HR 0.4, P < 0.001), and co-immunosuppression with corticosteroids (HR 0.64, P < 0.001) or azathioprine (HR 0.49, P < 0.001) were associated with impaired B-cell repopulation and reconstitution. Effects of corticosteroids (P = 0.043) and azathioprine (P = 0.025) were dose dependent. CONCLUSION Prolonged rituximab dosing intervals may be effective to achieve B-cell depletion and reduce risk of infection in advanced age or patients with impaired kidney function. Co-medication with corticosteroids or azathioprine prolongs B-cell recovery, which may increase therapeutic effects but also the rate of adverse events.
Collapse
Affiliation(s)
- Thomas Welte
- Department of Medicine IV, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lukas Westermann
- Department of Medicine IV, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Kappes
- Department of Pneumology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Markus A Schramm
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Xavier Bemtgen
- Department of Medicine III (Interdisciplinary Medical Intensive Care), Medical Center, Faculty of Medicine, University of Freiburg, and Department of Cardiology and Angiology I, Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dawid L Staudacher
- Department of Medicine III (Interdisciplinary Medical Intensive Care), Medical Center, Faculty of Medicine, University of Freiburg, and Department of Cardiology and Angiology I, Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin J Hug
- Pharmacy, Medical Center, University of Freiburg, Freiburg, Germany
| | - Nils Venhoff
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Frederic Arnold
- Department of Medicine IV, Medical Center, Faculty of Medicine, University of Freiburg, and Institute for Microbiology and Hygiene, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
5
|
Larsen Rasmussen S, Kumar P, Trebbien R, Leutscher P, Rasmussen C. Influenza Vaccine Type-Dependent Antibody Response in Patients with Autoimmune Inflammatory Rheumatic Diseases. Eur J Rheumatol 2023; 10:122-129. [PMID: 37873666 PMCID: PMC10765197 DOI: 10.5152/eurjrheum.2023.23053] [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: 06/16/2023] [Accepted: 08/31/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND The study aimed to explore influenza antibody response in patients with autoimmune inflammatory rheumatoid diseases (AIIRDs) stratified by the different vaccine types applied in Denmark during the 2018-2019 influenza season. METHODS Included patients were diagnosed with rheumatoid arthritis, psoriatic arthritis, or spondyloarthritis receiving biological disease-modifying antirheumatic drugs (bDMARDs) with or without conventional synthetic disease-modifying antirheumatic drugs. Influenza vaccination status in the 2018-2019 season and vaccine type received were reviewed in the Denmark. Blood samples were drawn ≥ 14 days post vaccination, and antibody titers were determined by the hemagglutinin inhibition (HAI) assay for the serotypes A/Michigan/H1N1, A/Singapore/H3N2, and B/Colorado included in the influenza vaccines in the 2018-2019 season. An overall serotype HAI geometric mean titer (GMT) was calculated from the 3 serotype-specific HAI titers. An overall serotype HAI GMT ≥ 40 was considered protective. RESULTS Of the 205 included patients, 105 (51%) had received influenza vaccination. One-quarter of vaccinated patients achieved post-vaccination overall serotype HAI GMT ≥40. For patients vaccinated with Influvac, a significantly higher proportion had HAI titers ≥ 40 for 2 serotypes, namely, A/Michigan/H1N1 and A/Singapore/H3N2, than patients vaccinated with Vaxigrip or VaxigripTetra. The same applied to all serotypes HAI GMT, where significantly more patients who received Influvac achieved postvaccination HAI GMT≥40 versus patients who received Vaxigrip (p=0.02) or VaxigripTetra (p=0.002). The latter outcome was explored in a multivariable logistic regression analysis and remained significant when including the following variables: age, sex, treatment with methotrexate and/or prednisolone, type of influenza vaccine, time interval from vaccination to antibody measurement, and previous vaccination status. CONCLUSION Influenza antibody levels following vaccination with Influvac in bDMARD-treated patients with AIIRDs were superior to Vaxigrip and VaxigripTetra. Treatment with methotrexate (MTX) did not reduce the antibody response.
Collapse
Affiliation(s)
| | - Prabhat Kumar
- Department of Rheumatology, North Denmark Regional Hospital, Hjoerring, Denmark
| | - Ramona Trebbien
- National Influenza Center, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Leutscher
- North Denmark Regional Hospital, Centre for Clinical Research, Hjoerring, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Claus Rasmussen
- North Denmark Regional Hospital, Centre for Clinical Research, Hjoerring, Denmark
- Department of Rheumatology, North Denmark Regional Hospital, Hjoerring, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
6
|
Clemente Garulo D, Núñez-Cuadros E, Camacho Lovillo M, Calzada-Hernández J, Guillén Martín S, Fernández Silveira L, Lirola Cruz MJ, Tagarro A, Alcobendas Rueda RM, López López A, Satrustegi Aritziturri M, Calvo C. Position statement on infection screening, prophylaxis, and vaccination in pediatric patients with rheumatic diseases and immunosuppressive therapies, part 2: infection prophylaxis. Eur J Pediatr 2023; 182:4271-4284. [PMID: 37439850 PMCID: PMC10570166 DOI: 10.1007/s00431-023-05080-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/14/2023]
Abstract
This study aims to provide practical recommendations on prophylaxis for infection in pediatric patients with immune-mediated rheumatic diseases receiving/scheduled to receive immunosuppressive therapy. A qualitative approach was applied. A narrative literature review was performed via Medline. Primary searches were conducted using MeSH terms and free text to identify articles that analyzed data on infections and vaccinations in pediatric patients with immune-mediated rheumatic diseases receiving immunosuppressive therapy. The results were presented and discussed in a nominal group meeting comprising a committee of 12 pediatric rheumatologists from the Prevention and Treatment of Infections Working Group of the Spanish Society of Pediatric Rheumatology. Several recommendations were generated. A consensus procedure was implemented via a Delphi process that was extended to members of the Spanish Society of Pediatric Rheumatology and the Vaccine Advisory Committee of the Spanish Association of Pediatrics. Participants produced a score ranging from 0 (completely disagree) to 10 (completely agree). Agreement was considered to have been reached if at least 70% of participants voted ≥ 7. The literature review included more than 400 articles. Overall, 63 recommendations were generated (23 on infection prophylaxis) and voted by 59 pediatric rheumatologists and other pediatric specialists, all of whom achieved the pre-established level of agreement. The recommendations on prophylaxis of infection cover vaccination and prophylaxis against varicella zoster virus, tuberculosis, Pneumocystis jiroveccii, and invasive fungal infections in pediatric patients with immune-mediated rheumatic diseases receiving/scheduled to receive immunosuppressive therapy. Conclusion: Based on current evidence and a Delphi process, we provided consensus and updated recommendations on prophylaxis and treatment of infections to guide those caring for pediatric rheumatology patients. What is Known: •Data largely derived from adults find that infectious diseases and related complications are a major cause of morbidity and mortality in patients with immune-mediated rheumatic diseases. •It is crucial to be aware of the preventive measures that should be implemented to prevent these infections in children, although most guidelines are often extrapolated from adult cases. What is New: •In the absence of evidence, a literature review and a Delphi survey were conducted to establish a series of expert recommendations that could prove useful in clinical practice, providing a practical and simple day-to-day approach to be used by pediatric rheumatologists. •The recommendations focus on tuberculosis, herpes zoster virus, fungal infections, and Pneumocystis jirovecii.
Collapse
Affiliation(s)
| | - Esmeralda Núñez-Cuadros
- Pediatric Rheumatology Unit, UGC Pediatría, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Av. del Arroyo de los Ángeles, S/N, 29011, Málaga, Spain.
| | - Marisol Camacho Lovillo
- Servicio de Inmunología, Hospital Universitario Virgen del Rocío/Instituto de Biomedicina de Sevilla (IBIS), Reumatología E Infectología Pediátricas, Seville, Spain
| | - Joan Calzada-Hernández
- Unitat de Reumatologia Pediàtrica, Servei de Pediatria, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Sara Guillén Martín
- Department of Paediatrics, Hospital Universitario de Getafe, Getafe, Madrid, Spain
| | - Laura Fernández Silveira
- Servicio de Inmunología, Hospital Universitario Virgen del Rocío/Instituto de Biomedicina de Sevilla (IBIS), Reumatología E Infectología Pediátricas, Seville, Spain
| | - María José Lirola Cruz
- Department of Paediatric Rheumatology, Instituto Hispalense de Pediatría, Seville, Spain
| | - Alfredo Tagarro
- Pediatrics Department, Instituto de Investigación 12 de Octubre (imas12), Hospital Universitario Infanta Sofía, Universidad Europea, Hospital 12 de Octubre, Madrid, Spain
| | | | - Agustín López López
- Department of Paediatrics, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | | | - Cristina Calvo
- Department of Pediatrics, Infectious and Tropical Diseases, Hospital Universitario La Paz, and La Paz Research Institute (IdiPaz), Madrid, Spain
- Translational Research Network of Pediatric Infectious Diseases (RITIP). Madrid, Madrid, Spain
| |
Collapse
|
7
|
Koehm M, Klippstein M, Dauth S, Hallmann K, Kohmer N, Burkhardt H, Ciesek S, Geisslinger G, Rabenau HF, Behrens F. Impact of different classes of immune-modulating treatments on B cell-related and T cell-related immune response before and after COVID-19 booster vaccination in patients with immune-mediated diseases and primary immunodeficiency: a cohort study. RMD Open 2023; 9:e003094. [PMID: 37652553 PMCID: PMC10476126 DOI: 10.1136/rmdopen-2023-003094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/23/2023] [Indexed: 09/02/2023] Open
Abstract
OBJECTIVES To evaluate the potential of immunosuppressed patients to mount B-cell and T-cell responses to COVID-19 booster vaccination (third vaccination). METHODS Patients with primary immunodeficiency (PID), immune-mediated inflammatory diseases (IMIDs) on CD20-depleting treatment with rituximab (RTX), or IMIDs treated with conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) or biological disease-modifying antirheumatic drug (bDMARDs) were included and assessed before (baseline visit (BL)) and 2, 4 and 8 weeks after COVID-19 booster vaccination. Serum B-cell responses were assessed by antibody levels against SARS-CoV-2 spike protein (anti-spike IgG antibody (S-AB)) and a surrogate virus neutralisation test (sVNT). T-cell responses were assessed by an interferon gamma release assay (IGRA). RESULTS Fifty patients with PID (n=6), treated with RTX therapy (n=13), or treated with csDMARDs/bDMARDs (n=31) were included. At BL, anti-S-AB titres in PID and csDMARD/bDMARD-treated patients were low (although significantly higher than RTX patients); measures of B-cell-mediated response increased significantly after booster vaccination. In the RTX cohort, low BL anti-S-AB and sVNT values did not improve after booster vaccination, but patients had significantly elevated IGRA responses post booster vaccination compared with the other groups. csDMARD/bDMARD-treated patients showed the highest BL values in all three assays with greater increases in all parameters after booster vaccination compared with patients with PID. CONCLUSION Patients with IMID on therapeutic B-cell depletion have low anti-S-AB and sVNT values before and after booster vaccination but show significantly higher levels of IGRA compared with other immunosuppressed patients, suggesting an underlying mechanism attempting to compensate compromised humoral immunity by upregulating T-cell responsiveness. PID appears to have a stronger impact on antiviral immune response than csDMARD/bDMARD treatment.
Collapse
Affiliation(s)
- Michaela Koehm
- Department of Rheumatology, Goethe University, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Maximilian Klippstein
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Stephanie Dauth
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Konstantin Hallmann
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Niko Kohmer
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Harald Burkhardt
- Department of Rheumatology, Goethe University, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Sandra Ciesek
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
- Virology, German Centre for Infection Research, Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Frankfurt am Main, Germany
| | - Holger F Rabenau
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Frank Behrens
- Department of Rheumatology, Goethe University, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| |
Collapse
|
8
|
Marcinkevics R, Silva PN, Hankele AK, Dörnte C, Kadelka S, Csik K, Godbersen S, Goga A, Hasenöhrl L, Hirschi P, Kabakci H, LaPierre MP, Mayrhofer J, Title AC, Shu X, Baiioud N, Bernal S, Dassisti L, Saenz-de-Juano MD, Schmidhauser M, Silvestrelli G, Ulbrich SZ, Ulbrich TJ, Wyss T, Stekhoven DJ, Al-Quaddoomi FS, Yu S, Binder M, Schultheiβ C, Zindel C, Kolling C, Goldhahn J, Seighalani BK, Zjablovskaja P, Hardung F, Schuster M, Richter A, Huang YJ, Lauer G, Baurmann H, Low JS, Vaqueirinho D, Jovic S, Piccoli L, Ciesek S, Vogt JE, Sallusto F, Stoffel M, Ulbrich SE. Machine learning analysis of humoral and cellular responses to SARS-CoV-2 infection in young adults. Front Immunol 2023; 14:1158905. [PMID: 37313411 PMCID: PMC10258347 DOI: 10.3389/fimmu.2023.1158905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 05/09/2023] [Indexed: 06/15/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces B and T cell responses, contributing to virus neutralization. In a cohort of 2,911 young adults, we identified 65 individuals who had an asymptomatic or mildly symptomatic SARS-CoV-2 infection and characterized their humoral and T cell responses to the Spike (S), Nucleocapsid (N) and Membrane (M) proteins. We found that previous infection induced CD4 T cells that vigorously responded to pools of peptides derived from the S and N proteins. By using statistical and machine learning models, we observed that the T cell response highly correlated with a compound titer of antibodies against the Receptor Binding Domain (RBD), S and N. However, while serum antibodies decayed over time, the cellular phenotype of these individuals remained stable over four months. Our computational analysis demonstrates that in young adults, asymptomatic and paucisymptomatic SARS-CoV-2 infections can induce robust and long-lasting CD4 T cell responses that exhibit slower decays than antibody titers. These observations imply that next-generation COVID-19 vaccines should be designed to induce stronger cellular responses to sustain the generation of potent neutralizing antibodies.
Collapse
Affiliation(s)
| | | | | | - Charlyn Dörnte
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Sarah Kadelka
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland
| | - Katharina Csik
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Svenja Godbersen
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Algera Goga
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Lynn Hasenöhrl
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Pascale Hirschi
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Hasan Kabakci
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Mary P. LaPierre
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Johanna Mayrhofer
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Xuan Shu
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Nouell Baiioud
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Zurich, Switzerland
| | - Sandra Bernal
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Zurich, Switzerland
| | - Laura Dassisti
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Meret Schmidhauser
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Zurich, Switzerland
| | - Giulia Silvestrelli
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Zurich, Switzerland
| | - Simon Z. Ulbrich
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Zurich, Switzerland
| | - Thea J. Ulbrich
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Zurich, Switzerland
| | - Tamara Wyss
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Zurich, Switzerland
| | - Daniel J. Stekhoven
- NEXUS Personalized Health Technologies, Zurich & SIB Swiss Institute of Bioinformatics, ETH Zurich, Lausanne, Switzerland
| | - Faisal S. Al-Quaddoomi
- NEXUS Personalized Health Technologies, Zurich & SIB Swiss Institute of Bioinformatics, ETH Zurich, Lausanne, Switzerland
| | - Shuqing Yu
- NEXUS Personalized Health Technologies, Zurich & SIB Swiss Institute of Bioinformatics, ETH Zurich, Lausanne, Switzerland
| | - Mascha Binder
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Christoph Schultheiβ
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Claudia Zindel
- Department of Health Science, Translational Medicine, ETH Zurich, Zurich, Switzerland
| | - Christoph Kolling
- Department of Health Science, Translational Medicine, ETH Zurich, Zurich, Switzerland
| | - Jörg Goldhahn
- Department of Health Science, Translational Medicine, ETH Zurich, Zurich, Switzerland
| | | | | | - Frank Hardung
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Marc Schuster
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Anne Richter
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Yi-Ju Huang
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Gereon Lauer
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | | | - Jun Siong Low
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Daniela Vaqueirinho
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Sandra Jovic
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Luca Piccoli
- Humabs BioMed SA, a Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Sandra Ciesek
- Institute of Medical Virology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Julia E. Vogt
- Department of Computer Science, ETH Zurich, Zurich, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
- Medical Immunology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Markus Stoffel
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
- University Hospital Zurich, Zurich, Switzerland
| | - Susanne E. Ulbrich
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
Azar JH, Evans JP, Sikorski MH, Chakravarthy KB, McKenney S, Carmody I, Zeng C, Teodorescu R, Song NJ, Hamon JL, Bucci D, Velegraki M, Bolyard C, Weller KP, Reisinger SA, Bhat SA, Maddocks KJ, Denlinger N, Epperla N, Gumina RJ, Vlasova AN, Oltz EM, Saif LJ, Chung D, Woyach JA, Shields PG, Liu SL, Li Z, Rubinstein MP. Selective suppression of de novo SARS-CoV-2 vaccine antibody responses in patients with cancer on B cell-targeted therapy. JCI Insight 2023; 8:e163434. [PMID: 36749632 PMCID: PMC10070099 DOI: 10.1172/jci.insight.163434] [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: 07/08/2022] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
We assessed vaccine-induced antibody responses to the SARS-CoV-2 ancestral virus and Omicron variant before and after booster immunization in 57 patients with B cell malignancies. Over one-third of vaccinated patients at the pre-booster time point were seronegative, and these patients were predominantly on active cancer therapies such as anti-CD20 monoclonal antibody. While booster immunization was able to induce detectable antibodies in a small fraction of seronegative patients, the overall booster benefit was disproportionately evident in patients already seropositive and not receiving active therapy. While ancestral virus- and Omicron variant-reactive antibody levels among individual patients were largely concordant, neutralizing antibodies against Omicron tended to be reduced. Interestingly, in all patients, including those unable to generate detectable antibodies against SARS-CoV-2 spike, we observed comparable levels of EBV- and influenza-reactive antibodies, demonstrating that B cell-targeting therapies primarily impair de novo but not preexisting antibody levels. These findings support rationale for vaccination before cancer treatment.
Collapse
Affiliation(s)
- Joseph H. Azar
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - John P. Evans
- Center for Retrovirus Research
- Department of Veterinary Biosciences
- Molecular, Cellular and Developmental Biology Program
| | - Madison H. Sikorski
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Karthik B. Chakravarthy
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Selah McKenney
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Ian Carmody
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Cong Zeng
- Center for Retrovirus Research
- Department of Veterinary Biosciences
| | - Rachael Teodorescu
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - No-Joon Song
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Jamie L. Hamon
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Donna Bucci
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Maria Velegraki
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Chelsea Bolyard
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Kevin P. Weller
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Sarah A. Reisinger
- The Ohio State University Comprehensive Cancer Center – James, The James Cancer Hospital
| | - Seema A. Bhat
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center – James
| | - Kami J. Maddocks
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center – James
| | - Nathan Denlinger
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center – James
| | - Narendranath Epperla
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center – James
| | - Richard J. Gumina
- Department of Internal Medicine, Division of Cardiovascular Medicine; and
| | - Anastasia N. Vlasova
- Center for Food Animal Health, Animal Sciences Department, Ohio Agricultural Research and Development Center, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Columbus, Ohio, USA
- Veterinary Preventive Medicine Department, College of Veterinary Medicine, The Ohio State University, Wooster, Ohio, USA
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute
| | - Eugene M. Oltz
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
- Department of Microbial Infection and Immunity; and
| | - Linda J. Saif
- Center for Food Animal Health, Animal Sciences Department, Ohio Agricultural Research and Development Center, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Columbus, Ohio, USA
- Veterinary Preventive Medicine Department, College of Veterinary Medicine, The Ohio State University, Wooster, Ohio, USA
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute
| | - Dongjun Chung
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio, USA
| | - Jennifer A. Woyach
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center – James
| | - Peter G. Shields
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Shan-Lu Liu
- Center for Retrovirus Research
- Department of Veterinary Biosciences
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute
- Department of Microbial Infection and Immunity; and
| | - Zihai Li
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Mark P. Rubinstein
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| |
Collapse
|
10
|
Bertozzi A, Mariottini A, Marchi L, Cristinzi MD, Nistri R, Damato V, Mechi C, Barilaro A, Massacesi L, Repice AM. Safety and effectiveness of the booster dose of mRNA COVID-19 vaccines in people with multiple sclerosis: A monocentric experience. Mult Scler Relat Disord 2023; 72:104582. [PMID: 36889098 PMCID: PMC9957336 DOI: 10.1016/j.msard.2023.104582] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/07/2023] [Accepted: 02/16/2023] [Indexed: 03/09/2023]
Abstract
BACKGROUND Long-term data on the effectiveness and safety of the booster dose of anti-SARS-CoV-2 vaccines in people affected by multiple sclerosis (pwMS) are lacking, hence a retrospective monocentric study exploring these issues was undertaken. MATERIALS AND METHODS PwMS who had received the booster dose of anti-COVID19 mRNA vaccines (either Comirnaty or Spikevax) according to the national regulation were included. The occurrence of adverse events or disease reactivation and SARS-CoV-2 infection were recorded up to last follow-up. Factors predictive of COVID-19 were explored using logistic regression analyses. A two-tailed p-value <0.05 was considered significant. RESULTS One hundred and fourteen pwMS were included: 80 females (70%); median age at the booster dose 42 years (range 21 - 73); 106/114 patients (93%) were receiving a disease-modifying treatment at vaccination. The median follow-up after the booster dose was 6 (range 2 - 7) months. Adverse events were experienced in 58% of the patients, being mild to moderate in most cases; 4 reactivations of MS were observed, two of which occurring within 4 weeks after the booster. SARS-CoV-2 infection was reported in 24/114 (21%) cases, occurring a median of 74 days (5-162) after the booster dose and requiring hospitalisation in 2 patients. Six cases received direct antiviral drugs. Age at vaccination and time between the primary vaccination cycle and the booster dose were independently and inversely associated with the risk of COVID-19 (HR 0.95 and 0.98, respectively). CONCLUSIONS The administration of the booster dose in pwMS showed an overall good safety profile and protected 79% of the patients from SARS-CoV-2 infection. The observed association between the risk of infection after the booster dose and both younger age at vaccination and shorter interval period to the booster dose suggest that unobserved confounders, possibly including behavioural and social factors, play a relevant role in determining the individual propensity to get infected with COVID-19.
Collapse
Affiliation(s)
- Andrea Bertozzi
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy
| | - Alice Mariottini
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy; Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Centre, Careggi University Hospital, Florence, Italy.
| | - Leonardo Marchi
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy
| | - Maria Di Cristinzi
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy
| | - Riccardo Nistri
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy; University "La Sapienza", Rome, Italy
| | - Valentina Damato
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy; Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Centre, Careggi University Hospital, Florence, Italy
| | - Claudia Mechi
- Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Centre, Careggi University Hospital, Florence, Italy
| | - Alessandro Barilaro
- Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Centre, Careggi University Hospital, Florence, Italy
| | - Luca Massacesi
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy; Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Centre, Careggi University Hospital, Florence, Italy
| | - Anna Maria Repice
- Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Centre, Careggi University Hospital, Florence, Italy
| |
Collapse
|
11
|
Ren J, Wang J, Liu R, Guo J, Yao Y, Luo J, Hao H, Gao F. Case report: Persistent hypogammaglobulinemia in thymoma-associated myasthenia gravis: the impact of rituximab or Good's syndrome? Front Neurol 2023; 14:1152992. [PMID: 37213908 PMCID: PMC10196171 DOI: 10.3389/fneur.2023.1152992] [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: 02/01/2023] [Accepted: 04/12/2023] [Indexed: 05/23/2023] Open
Abstract
Introduction Rituximab (RTX) showed good efficacy and safety for patients with myasthenia gravis. However, the percentage of peripheral CD20+ B cell may be absent for years after low dose of RTX treatment. Persistent hypogammaglobulinemia and opportunistic infection may occur in patients under treatment of RTX with thymoma relapse. Case representation We report a case of refractory myasthenia gravis. After two doses of 100 mg rituximab, the patient developed transient neutropenia. The peripheral blood CD20+ B cell percentage was 0 more than 3 years. Eighteen months later, the patient's symptoms relapsed with thymoma recurred. She had persistent hypogammaglobulinemia and multiple opportunistic infections. Conclusion In MG patient under B cell depletion therapy had thymoma relapse, Good's syndrome may induce prolonged B cell depletion, hypogammaglobulinemia and opportunistic infections.
Collapse
|
12
|
El Sammak DAEA, Abdelhay RM. Role of [18F] FDG PET-CT in detection of COVID-19 vaccine-associated hypermetabolic lymphadenopathy (VAHL) in lymphoma patients: with serologic testing correlation. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2023; 54:26. [PMCID: PMC9893975 DOI: 10.1186/s43055-022-00896-9] [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: 02/05/2023] Open
Abstract
Background COVID-19 vaccination of the population has a great importance, especially in oncological patients. The high incidence of vaccine-associated hypermetabolic lymphadenopathy (VAHL) makes a difficulty in the diagnosis of PET-CT of oncological patients. They should be vaccinated in the side opposite to the expected malignant LNs to avoid unnecessary biopsy and change in therapy. The aim of this study was to assess the role of PET-CT in detection of VAHL after the 2nd dose of Pfizer-BioNTech vaccine in lymphoma patients and compare the incidence of VAHL among lymphoma patients treated with B cell depletion therapy during the 6 months prior to vaccination and those treated > 6 months before vaccination. Results This study comprised 120 lymphoma patients, referred for FDG PET/CT 1–3 weeks after the 2nd dose of Pfizer-BioNTech COVID-19 vaccine. Hypermetabolic LNs were identified in 55%. The incidence of VAHL in lymphoma patients treated with anti-CD20 antibody rituximab during the 6 months prior to vaccination (9%) was significantly lower compared with other lymphoma patients treated with anti-CD20 antibody rituximab > 6 months before vaccination (91%). The incidence and grades of VAHL are significantly high within the 1st week after the 2nd dose of Pfizer-BioNTech vaccine in patients younger than 60 years of age. Only 7 of 37 patients with negative serology had VAHL on PET-CT, whereas 10 of 26 patients with decreased anti-spike titers and 49 of 57 patients with increased anti-spike titers had VAHL on PET-CT. Conclusions VAHL makes challenges in the interpretation of FDG PET/CT in oncology patients. Accurate data collection, regarding the time and site of COVID vaccination, is important to help radiologists in identifying the cause of abnormal nodal FDG uptake. We suggest to schedule FDG PET-CT for lymphoma patients at least 3 weeks after the 2nd dose of Pfizer-BioNTech vaccine.
Collapse
Affiliation(s)
- Dena Abd El Aziz El Sammak
- grid.31451.320000 0001 2158 2757Egypt Radiodiagnosis Department, Zagazig University Hospital, Zagazig, Egypt
| | - Rabab M. Abdelhay
- grid.31451.320000 0001 2158 2757Radiodiagnosis Department, Zagazig University Hospital, Egypt, Zagazig, Egypt
| |
Collapse
|
13
|
Shimizu A, Shirai I, Ogawa K, Miura A, Haruhara K, Oshiro K, Hamaguchi A, Yokote S, Okabe M, Ueda H, Tsuboi N, Ikeda M, Yokoo T. Persistent SARS-CoV-2 Infection in a Patient with Nephrotic Syndrome under Rituximab Therapy: Successful Treatment with a Combination of Remdesivir and Monoclonal Antibodies. Intern Med 2022; 61:3703-3708. [PMID: 36171121 PMCID: PMC9841105 DOI: 10.2169/internalmedicine.0241-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Rituximab is an effective treatment for frequently relapsing/steroid-dependent nephrotic syndrome, but there is concern about infections caused by humoral immunodeficiency. We herein report a case of prolonged (>7 weeks) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. A 24-year-old man with minimal change disease treated with rituximab developed SARS-CoV-2 infection. The clinical response to remdesivir was soon transiently abolished. Treatment with casirivimab and imdevimab (REGEN-COV) monoclonal antibodies in combination with remdesivir resulted in complete clearance of the infection. The REGEN-COV antibody cocktail may improve the outcome of SARS-CoV-2 infection in patients with humoral immunodeficiency.
Collapse
Affiliation(s)
- Akihiro Shimizu
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Japan
| | - Izumi Shirai
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Japan
| | - Kyohei Ogawa
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Japan
| | - Akane Miura
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Japan
| | - Kotaro Haruhara
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Japan
| | - Kentaro Oshiro
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Japan
| | - Akihiko Hamaguchi
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Japan
| | - Shinya Yokote
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University Katsushika Medical Center, Japan
| | - Masahiro Okabe
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University Daisan Hospital, Japan
| | - Hiroyuki Ueda
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Japan
| | - Nobuo Tsuboi
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Japan
| | - Masato Ikeda
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Japan
| |
Collapse
|
14
|
Sood A, Tran M, Murthy V, Gonzalez E. Immunogenicity and Safety of SARS-CoV-2 Vaccination in Patients With Rheumatic Diseases: A Systematic Review and Meta-analysis. J Clin Rheumatol 2022; 28:381-389. [PMID: 35660717 DOI: 10.1097/rhu.0000000000001871] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE The aim of this study was to conduct a systematic review and meta-analysis examining the immunogenicity and safety of SARS-CoV-2 vaccination in patients with RD. METHODS We systematically searched PubMed/MEDLINE and Scopus to identify observational studies that examined the immunogenicity and safety of SARS-CoV-2 vaccination in RD patients. Information on disease, immunosuppressant, vaccine type, and proportion of patients with serologic response was obtained from each study. RESULTS There were 25 eligible studies. The pooled rate of seroconversion was 0.79 (95% confidence interval [CI], 0.72-0.86). Compared with control subjects, the odds of seroconversion were significantly lower (odds ratio, 0.11; 95% CI, 0.05-0.24). Users of rituximab showed the lowest rate of seroconversion (0.39; 95% CI, 0.29-0.51) followed by mycophenolate (0.56; 95% CI, 0.40-71). On the other hand, users of interleukin 17 (0.94; 95% CI, 0.78-0.98) and tumor necrosis factor inhibitors (0.94; 95% CI, 0.84-0.98) showed high seroconversion rate. Regarding safety of COVID-19 vaccine, approximately 2% of patients reported severe adverse events and 7% reported diseases flares following the first or second dose. CONCLUSION Vaccination against SARS-CoV-2 appears to be safe. Most RD patients developed humoral immune response following vaccination. However, the odds of seroconversion were significantly lower in RD patients compared with controls. This is likely driven by certain immunosuppressants including rituximab and mycophenolate. Future studies need to identify strategies to improve vaccine response in these patients.
Collapse
Affiliation(s)
- Akhil Sood
- From the Departments of Internal Medicine
| | - Minh Tran
- From the Departments of Internal Medicine
| | - Vijaya Murthy
- Rheumatology, University of Texas Medical Branch, Galveston, TX
| | - Emilio Gonzalez
- Rheumatology, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
15
|
Torres M, Corona M, Rodríguez-Mora S, Casado-Fernández G, Zurdo-Castronuño A, Mateos E, Ramos-Martín F, Sánchez-Menéndez C, Murciano-Antón MA, García-Pérez J, Alcamí J, Pérez-Olmeda M, Coiras M, López-Jiménez J, García-Gutiérrez V. Strong Humoral but Not Cellular Immune Responses against SARS-CoV-2 in Individuals with Oncohematological Disease Who Were Treated with Rituximab before Receiving a Vaccine Booster. Cancers (Basel) 2022; 14:5537. [PMID: 36428631 PMCID: PMC9688562 DOI: 10.3390/cancers14225537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/03/2022] [Accepted: 11/06/2022] [Indexed: 11/12/2022] Open
Abstract
The humoral immune response developed after receiving the full vaccination schedule against COVID-19 is impaired in individuals who received anti-CD20 therapy 6-9 months before vaccination. However, there is little information about the cellular immune responses elicited in these individuals. In this study, we analyzed the humoral and cellular immune responses in 18 individuals with hematological disease who received the last dose of rituximab 13.8 months (IQR 9.4-19) before the booster dose. One month after receiving the booster dose, the seroconversion rate in the rituximab-treated cohort increased from 83.3% to 88.9% and titers of specific IgGs against SARS-CoV-2 increased 1.53-fold (p = 0.0098), while the levels of neutralizing antibodies increased 3.03-fold (p = 0.0381). However, the cytotoxic activity of peripheral blood mononuclear cells (PBMCs) from rituximab-treated individuals remained unchanged, and both antibody-dependent cellular cytotoxicity (ADCC) and direct cellular cytotoxicity (CDD) were reduced 1.7-fold (p = 0.0047) and 2.0-fold (p = 0.0086), respectively, in comparison with healthy donors. Breakthrough infections rate was higher in our cohort of rituximab-treated individuals (33.33%), although most of the infected patients (83.4%) developed a mild form of COVID-19. In conclusion, our findings confirm a benefit in the humoral, but not in the cellular, immune response in rituximab-treated individuals after receiving a booster dose of an mRNA-based vaccine against COVID-19.
Collapse
Affiliation(s)
- Montserrat Torres
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Magdalena Corona
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Faculty of Sciences, Universidad de Alcalá, 28871 Alcalá de Henares, Spain
| | - Sara Rodríguez-Mora
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | - Guiomar Casado-Fernández
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Faculty of Sciences, Universidad de Alcalá, 28871 Alcalá de Henares, Spain
| | - Alejandro Zurdo-Castronuño
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Elena Mateos
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | - Fernando Ramos-Martín
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | - Clara Sánchez-Menéndez
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | | | - Javier García-Pérez
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28222 Majadahonda, Spain
| | - José Alcamí
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28222 Majadahonda, Spain
| | - Mayte Pérez-Olmeda
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Serology Service, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mayte Coiras
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | - Javier López-Jiménez
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Valentín García-Gutiérrez
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | | |
Collapse
|
16
|
Advancing Biologic Therapy for Refractory Autoimmune Hepatitis. Dig Dis Sci 2022; 67:4979-5005. [PMID: 35147819 DOI: 10.1007/s10620-021-07378-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/27/2021] [Indexed: 01/05/2023]
Abstract
Biologic agents may satisfy an unmet clinical need for treatment of refractory autoimmune hepatitis. The goals of this review are to present the types and results of biologic therapy for refractory autoimmune hepatitis, indicate opportunities to improve and expand biologic treatment, and encourage comparative clinical trials. English abstracts were identified in PubMed by multiple search terms. Full-length articles were selected for review, and secondary and tertiary bibliographies were developed. Rituximab (monoclonal antibodies against CD20 on B cells), infliximab (monoclonal antibodies against tumor necrosis factor-alpha), low-dose recombinant interleukin 2 (regulatory T cell promoter), and belimumab (monoclonal antibodies against B cell activating factor) have induced laboratory improvement in small cohorts with refractory autoimmune hepatitis. Ianalumab (monoclonal antibodies against the receptor for B cell activating factor) is in clinical trial. These agents target critical pathogenic pathways, but they may also have serious side effects. Blockade of the B cell activating factor or its receptors may disrupt pivotal B and T cell responses, and recombinant interleukin 2 complexed with certain interleukin 2 antibodies may selectively expand the regulatory T cell population. A proliferation-inducing ligand that enhances T cell proliferation and survival is an unevaluated, potentially pivotal, therapeutic target. Fully human antibodies, expanded target options, improved targeting precision, more effective delivery systems, and biosimilar agents promise to improve efficacy, safety, and accessibility. In conclusion, biologic agents target key pathogenic pathways in autoimmune hepatitis, and early experiences in refractory disease encourage clarification of the preferred target, rigorous clinical trial, and comparative evaluations.
Collapse
|
17
|
Terada M, Kondo N, Wanifuchi-Endo Y, Fujita T, Asano T, Hisada T, Uemoto Y, Akiko Kato, Yamanaka N, Sugiura H, Mita K, Wada A, Takahashi E, Saito K, Yoshioka R, Toyama T. Efficacy and impact of SARS-CoV-2 vaccination on cancer treatment for breast cancer patients: a multi-center prospective observational study. Breast Cancer Res Treat 2022; 195:311-323. [PMID: 35941421 PMCID: PMC9360656 DOI: 10.1007/s10549-022-06693-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/20/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE Vaccination is an essential strategy to prevent infection in the SARS-CoV-2 pandemic. However, there are concerns about vaccine efficacy and the impact of vaccination on cancer treatment. Additionally, the emergence of novel variants may affect vaccination efficacy. This multi-center, prospective, observational study investigated the efficacy and impact of vaccination against SARS-CoV-2 variants on treatment among breast cancer patients in Japan. METHODS Patients with breast cancer scheduled to be vaccinated with the SARS-CoV-2 vaccine from May to November 2021 were prospectively enrolled (UMIN000045527). They were stratified into five groups according to their cancer treatment: no treatment, hormone therapy, anti-human epidermal growth factor receptor (HER)2 therapy, chemotherapy, and cyclin-dependent kinase 4/6 (CDK4/6) inhibitor. Serum samples for assessing serological responses were collected before the first vaccination and after the second vaccination. RESULTS Eighty-five breast cancer patients were included. The overall seroconversion rate after second vaccination was 95.3% and the lowest seroconversion rate was 81.8% in the patients under chemotherapy. The overall positivity rate of neutralizing antibodies against the wild-type, α, Δ, κ, and omicron variants were 90.2%, 81.7%, 96.3%, 84.1%, and 8.5%, respectively. Among the patients under chemotherapy or CDK4/6 inhibitors, various degrees of decreased neutralizing antibody titers against SARS-CoV-2 variants were observed. Withdrawal or reduction of systemic therapy because of vaccination was observed in only one patient. CONCLUSION Our data support SARS-CoV-2 vaccination for breast cancer patients. However, a reduction in neutralizing antibody titers was suggested during chemotherapy and CDK4/6 inhibitors, raising concerns about the impact on long-term infection prevention.
Collapse
Affiliation(s)
- Mitsuo Terada
- Departments of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Naoto Kondo
- Departments of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Yumi Wanifuchi-Endo
- Departments of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Takashi Fujita
- Departments of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Tomoko Asano
- Departments of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Tomoka Hisada
- Departments of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Yasuaki Uemoto
- Departments of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Akiko Kato
- Departments of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Natsumi Yamanaka
- Departments of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Hiroshi Sugiura
- Departments of Breast and Endocrine Surgery, Nagoya City University West Medical Center, 1-1-1 Hirate-cho, Kita-ku, Nagoya, Aichi, 462-8508, Japan
| | - Keiko Mita
- Departments of Breast and Endocrine Surgery, Nagoya City University East Medical Center, 1-2-23 Wakamizu, Chikusa-ku, Nagoya, Aichi, 464-8547, Japan
| | - Asaka Wada
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University, S1W16, Chuoh-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Eriko Takahashi
- Department of Breast and Endocrine Surgery, Akita University Hospital, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Kanako Saito
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Ryo Yoshioka
- Department of General Thoracic Surgery and Breast and Endocrine Surgery, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Tatsuya Toyama
- Departments of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan.
| |
Collapse
|
18
|
Tang K, Wei Z, Wu X. Impaired serological response to COVID-19 vaccination following anti-cancer therapy: a systematic review and meta-analysis. J Med Virol 2022; 94:4860-4868. [PMID: 35750492 PMCID: PMC9349696 DOI: 10.1002/jmv.27956] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 11/25/2022]
Abstract
Owing to the high coronavirus disease 2019 (COVID‐19)‐related morbidity and fatality rate among patients with cancer, the introduction of COVID‐19 vaccines is of profound significance in this fragile population. Accumulating data suggested that oncologic patients, especially those with anticancer therapy have an impaired immune response to COVID‐19 vaccination. However, the exact effect of anticancer treatments on postvaccination response has not been elucidated yet. We, therefore, conducted a meta‐analysis to evaluate the impact of treatments on response to COVID‐19 vaccination in patients with cancer. A total of 39 studies were finally included comprising 11 075 oncologic patients. Overall, we found the humoral response was significantly decreased in patients undergoing anticancer treatments (odds ratio [OR] = 2.55, 95% confidence interval [CI]: 2.04–3.18) compared with those without active treatment. The seroconversion rates were significantly lower in patients with chemotherapy (OR = 3.04, 95% CI: 2.28–4.05), targeted therapy (OR = 4.72, 95% CI: 3.18–7.01) and steroid usage (OR = 2.19, 95% CI: 1.57–3.07), while there was no significant association between immunotherapy or hormonal therapy and seroconversion after vaccination. Subgroup analyses showed therapies with anti‐CD20 antibody (OR = 11.28, 95% CI: 6.40–19.90), B‐cell lymphoma 2 inhibitor (OR = 5.76, 95% CI: 3.64–9.10), and Bruton tyrosine kinase inhibitor (OR = 6.86, 95% CI: 4.23–11.15) were significantly correlated with the risk of negative humoral response to vaccination. In conclusion, our results demonstrated that specific oncologic therapies may significantly affect serological response to COVID‐19 vaccines in patients with cancer. Thus, an adapted vaccination strategy taking the influence of active treatment into account is in need, and further research on the effect of the third dose of vaccine and the role of postvaccination cellular response in oncologic patients is also needed.
Collapse
Affiliation(s)
- Kefu Tang
- Prenatal Diagnosis Center, Department of Clinical Laboratory, Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, 200051, China
| | - Zhiying Wei
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Xi Wu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University,, Shanghai, 200030, China
| |
Collapse
|
19
|
Martire B, Ottaviano G, Sangerardi M, Sgrulletti M, Chini L, Dellepiane RM, Montin D, Rizzo C, Pignata C, Marseglia GL, Moschese V. Vaccinations in Children and Adolescents Treated With Immune-Modifying Biologics: Update and Current Developments. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:1485-1496. [PMID: 35085809 DOI: 10.1016/j.jaip.2022.01.007] [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: 07/02/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 06/14/2023]
Abstract
Treatment with immune-modifying biologics has positively impacted disease control and quality of life in many patients with immune-mediated disorders. However, the higher susceptibility to common and opportunistic pathogens is of concern. Thus, immunization strategies to control vaccine-preventable diseases represent a critical issue in this population. However, limited data exist on the safety, immunogenicity, and efficacy of available vaccines in patients on biologics, particularly in children. Here, according to published literature and real-life experience and practice, we report the interim indications of the Italian Society of Pediatric Allergology and Immunology (SIAIP) Vaccine Committee and of the Italian Primary Immunodeficiency Network (IPINet) Centers on immunization of children and adolescents receiving biologics. Our aim is to provide a practical guidance for the clinician to ensure optimal protection for patients and the community.
Collapse
Affiliation(s)
- Baldassarre Martire
- Pediatrics and Neonatology Unit, Maternal-Infant Department, Monsignor A. R. Dimiccoli Hospital, Barletta, Italy.
| | - Giorgio Ottaviano
- Molecular and Cellular Immunology Unit, Great Ormond Street Institute of Child Health, University College of London, London, UK
| | - Maria Sangerardi
- Department of Pediatrics and Emergency, Pediatric Hospital, Policlinico - University of Bari, Bari, Italy
| | - Mayla Sgrulletti
- Pediatric Immunopathology and Allergology Unit, University of Rome, Tor Vergata, Policlinico Tor Vergata, Rome, Italy
| | - Loredana Chini
- Pediatric Immunopathology and Allergology Unit, University of Rome, Tor Vergata, Policlinico Tor Vergata, Rome, Italy
| | - Rosa Maria Dellepiane
- Pediatric Intermediate Care Unit, Scientific Institute for Research, Hospitalization and Healthcare Foundation (IRCSS); Ca Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Davide Montin
- Department of Public Health and Pediatrics, Regina Margherita Children Hospital, University of Turin, Turin, Italy
| | - Caterina Rizzo
- Innovation and Clinical Pathways Unit, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Claudio Pignata
- Department of Translational Medical Sciences-Section of Pediatrics, Federico II University of Naples, Naples, Italy
| | - Gian Luigi Marseglia
- Department of Pediatrics, IRCCS Foundation, Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Viviana Moschese
- Pediatric Immunopathology and Allergology Unit, University of Rome, Tor Vergata, Policlinico Tor Vergata, Rome, Italy
| |
Collapse
|
20
|
Fujii T, Hagihara M, Mitamura K, Nakashima S, Ohara S, Uchida T, Inoue M, Okuda M, Yasuhara A, Murakami J, Duong C, Iwatsuki-Horimoto K, Yamayoshi S, Kawaoka Y. Anti-SARS CoV-2 IgG in COVID-19 Patients with Hematological Diseases: A Single-center, Retrospective Study in Japan. Intern Med 2022; 61:1681-1686. [PMID: 35342138 PMCID: PMC9259303 DOI: 10.2169/internalmedicine.9209-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Objective Coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread globally. Although the relationship between anti-SARS-CoV-2 immunoglobulin G (IgG) antibodies and COVID-19 severity has been reported, information is lacking regarding the seropositivity of patients with particular types of diseases, including hematological diseases. Methods In this single-center, retrospective study, we compared SARS-CoV-2 IgG positivity between patients with hematological diseases and those with non-hematological diseases. Results In total, 77 adult COVID-19 patients were enrolled. Of these, 30 had hematological disorders, and 47 had non-hematological disorders. The IgG antibody against the receptor-binding domain of the spike protein was detected less frequently in patients with hematological diseases (60.0%) than in those with non-hematological diseases (91.5%; p=0.029). Rituximab use was significantly associated with seronegativity (p=0.010). Conclusion Patients with hematological diseases are less likely to develop anti-SARS-CoV-2 antibodies than those with non-hematological diseases, which may explain the poor outcomes of COVID-19 patients in this high-risk group.
Collapse
Affiliation(s)
- Takayuki Fujii
- Department of Hematology, Eiju General Hospital, Japan
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Japan
| | | | - Keiko Mitamura
- Division of Infection Control, Eiju General Hospital, Japan
| | | | - Shin Ohara
- Department of Hematology, Eiju General Hospital, Japan
| | | | | | - Moe Okuda
- Division of Virology, Institute of Medical Science, University of Tokyo, Japan
| | - Atsuhiro Yasuhara
- Division of Virology, Institute of Medical Science, University of Tokyo, Japan
| | - Jurika Murakami
- Division of Virology, Institute of Medical Science, University of Tokyo, Japan
| | - Calvin Duong
- Division of Virology, Institute of Medical Science, University of Tokyo, Japan
| | | | - Seiya Yamayoshi
- Division of Virology, Institute of Medical Science, University of Tokyo, Japan
- The Research Center for Global Viral Diseases, Research Institute, National Center for Global Health and Medicine, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Institute of Medical Science, University of Tokyo, Japan
- The Research Center for Global Viral Diseases, Research Institute, National Center for Global Health and Medicine, Japan
| |
Collapse
|
21
|
Tolf A, Wiberg A, Müller M, Nazir FH, Pavlovic I, Laurén I, Mangsbo S, Burman J. Factors Associated With Serological Response to SARS-CoV-2 Vaccination in Patients With Multiple Sclerosis Treated With Rituximab. JAMA Netw Open 2022; 5:e2211497. [PMID: 35544139 PMCID: PMC9096596 DOI: 10.1001/jamanetworkopen.2022.11497] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/24/2022] [Indexed: 12/14/2022] Open
Abstract
Importance B-cell-depleting monoclonal antibodies are widely used for treatment of multiple sclerosis but are associated with an impaired response to vaccines. Objective To identify factors associated with a favorable vaccine response to tozinameran. Design, Setting, and Participants This prospective cohort study was conducted in a specialized multiple sclerosis clinic at a university hospital from January 21 to December 1, 2021. Of 75 patients evaluated for participation who received a diagnosis of multiple sclerosis with planned or ongoing treatment with rituximab, 69 were included in the study, and data from 67 were analyzed. Exposures Sex, age, number of previous rituximab infusions, accumulated dose of rituximab, previous COVID-19 infection, time since last rituximab treatment, CD19+ B-cell count before vaccination, CD4+ T-cell count, and CD8+ T-cell count were considered potential factors associated with the main outcome. Main Outcomes and Measures Serological vaccine responses were measured by quantitation of anti-spike immunoglobulin G (IgG) antibodies, anti-receptor-binding domain (RBD) IgG antibodies, and their neutralizing capacities. Cellular responses to spike protein-derived SARS-CoV-2 peptide pools were assessed by counting interferon gamma spot-forming units in a FluoroSpot assay. Results Among 60 patients with ongoing rituximab treatment (49 women [82%]; mean (SD) age, 43 [10] years), the median (range) disease duration was 9 (1-29) years, and the median (range) dose of rituximab was 2750 (500-10 000) mg during a median (range) time of 2.8 (0.5-8.3) years. The median (range) follow-up from the first vaccination dose was 7.3 (4.3-10.0) months. Vaccine responses were determined before vaccination with tozinameran and 6 weeks after vaccination. By using established cutoff values for anti-spike IgG (264 binding antibody units/mL) and anti-RBD IgG (506 binding antibody units/mL), the proportion of patients with a positive response increased with the number of B cells, which was the only factor associated with these outcomes. A cutoff for the B-cell count of at least 40/μL was associated with an optimal serological response. At this cutoff, 26 of 29 patients (90%) had positive test results for anti-spike IgG and 21 of 29 patients (72%) for anti-RBD IgG, and 27 of 29 patients (93%) developed antibodies with greater than 90% inhibition of angiotensin-converting enzyme 2. No factor associated with the cellular response was identified. Depending on the peptide pool, 21 of 25 patients (84%) to 22 of 25 patients (88%) developed a T-cell response with interferon gamma production at the B-cell count cutoff of at least 40/μL. Conclusions and Relevance This cohort study found that for an optimal vaccine response from tozinameran, rituximab-treated patients with multiple sclerosis may be vaccinated as soon as possible, with rituximab treatment delayed until B-cell counts have reached at least 40/μL. An additional vaccination with tozinameran should be considered at that point.
Collapse
Affiliation(s)
- Andreas Tolf
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Anna Wiberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Malin Müller
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Ivan Pavlovic
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Ida Laurén
- Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Sara Mangsbo
- Department of Pharmacy, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Joachim Burman
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
22
|
Ishida H, Furusawa M, Unagami K, Omoto K, Iizuka J, Takagi T. Antibody Response to SARS-CoV-2 mRNA Vaccine Among Kidney Transplant Recipients: A Retrospective Cohort Study at a Single Transplant Institute in Japan. EXP CLIN TRANSPLANT 2022; 20:463-471. [PMID: 35607795 DOI: 10.6002/ect.2022.0020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Although the effectiveness of vaccines in protecting the host from infection has been proven, few surveys have been conducted on changes in antibody levels after vaccination of kidney transplant recipients in Japan. MATERIALS AND METHODS We analyzed serological responses in kidney transplant recipients after BNT162B2 COVID-19 mRNA vaccine with the use of a reagent capable of simultaneously specifying the antibody response to 5 proteins: a full-spike protein (extracellular domain), 3 individual domains of the spike protein (S1, S2, and receptor-binding domain), and nucleocapsid. The analysis involved 111 patients who had follow-up over 1 month after having received the second of 2 coronavirus vaccines after kidney transplant. RESULTS Antibodies were detected in 46 of 111 patients (41%). The antibody-positive rate in the kidney transplant group tended to be lower than that in the healthy control group, which showed an antibody- positive rate of 100%. When the antibody-positive rate was analyzed by the type of immunosuppressor used, the rate was 36% (37/100) for patients who used tacrolimus at the time of vaccination and 90% (9/10) for patients who used cyclosporine. Patients administered CD20 antibody (rituximab) before and/or after transplant showed a lower production of antibodies, which was supported by a smaller number of CD19- and CD20-positive cells in the peripheral blood as well as a shorter period between rituximab administration and vaccination. The percentage of responding viral fragments varied greatly among individual patients and showed no uniformity in the kidney transplant group, whereas the mean fluorescence intensity of individual fragments showed a certain tendency in the control group. CONCLUSIONS The appropriate timing of vaccination should be considered in transplant recipients who use tacrolimus-mycophenolate mofetil combination and rituximab as these drugs are deeply related to a lower antibody response to SARS-CoV-2 BNT162b2 vaccination.
Collapse
Affiliation(s)
- Hideki Ishida
- From the Department of Urology, , Women's Medical University Hospital, Tokyo, Japan.,From the Department of Transplant Medicine, Women's Medical University Hospital, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
23
|
Lee ARYB, Wong SY, Tay SH. Booster COVID-19 Vaccines for Immune-Mediated Inflammatory Disease Patients: A Systematic Review and Meta-Analysis of Efficacy and Safety. Vaccines (Basel) 2022; 10:668. [PMID: 35632424 PMCID: PMC9144569 DOI: 10.3390/vaccines10050668] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Seroconversion and longevity of vaccine-induced immune response is blunted in immune-mediated inflammatory disease (IMID) patients owing to immunosuppressive regimens. COVID-19 booster vaccines after a primary series have been proposed with inconclusive evidence on efficacy to date. METHODS This PROSPERO-registered systematic review (CRD42022302534) was conducted according to PRISMA guidelines. PubMed, EMBASE, CENTRAL, Web of Science, CORD-19, WHO ICTRP, and medRxiv were searched up to 28 February 2022 for eligible studies. Risk of bias was assessed using the Joanna Briggs Institute critical appraisal tools. RESULTS From 6647 records, 17 prospective studies were included for systematic review and 12 in meta-analysis of primary series non-responders. The risk of bias was low. Pooling 340 non-responders, a booster dose proved effective with 0.47 seroconverting (95% CI: 0.32-0.63, I2 = 82%). Rituximab therapy was associated with significant impairment, with risks of 0.25 (95% CI: 0.17-0.36, I2 = 50.7%) versus 0.81 (95% CI: 0.72-0.87, I2 = 0.0%) for those without rituximab therapy. A systematic review of antibody levels against COVID-19 showed several-fold increases across studies. Incidence of local and systemic adverse events, including disease flares, were either comparable or slightly increased after the booster dose compared to primary series. No major events such as myocarditis or death were reported. CONCLUSION Our results show that booster doses are effective in eliciting seroconversion in non-responders, bolstering immunity to COVID-19. It has also not been associated with major adverse events.
Collapse
Affiliation(s)
- Ainsley Ryan Yan Bin Lee
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (A.R.Y.B.L.); (S.Y.W.)
| | - Shi Yin Wong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (A.R.Y.B.L.); (S.Y.W.)
| | - Sen Hee Tay
- Division of Rheumatology, Department of Medicine, National University Hospital, Singapore 119228, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| |
Collapse
|
24
|
Bemben NM, Berg ML. Efficacy of inactivated vaccines in patients treated with immunosuppressive drug therapy. Pharmacotherapy 2022; 42:334-342. [PMID: 35146780 PMCID: PMC9088666 DOI: 10.1002/phar.2671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 12/17/2022]
Affiliation(s)
- Nina M. Bemben
- Wolters Kluwer Clinical Effectiveness Chicago Illinois USA
| | - Melody L. Berg
- American Society of Health‐System Pharmacists Bethesda Maryland USA
| |
Collapse
|
25
|
Bacova B, Kohutova Z, Zubata I, Gaherova L, Kucera P, Heizer T, Mikesova M, Karel T, Novak J. Cellular and humoral immune response to SARS-CoV-2 mRNA vaccines in patients treated with either Ibrutinib or Rituximab. Clin Exp Med 2022; 23:371-379. [PMID: 35352210 PMCID: PMC8963888 DOI: 10.1007/s10238-022-00809-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 02/05/2022] [Indexed: 01/05/2023]
Abstract
Patients treated with B-cell-targeting therapies like Rituximab or Ibrutinib have decreased serological response to various vaccines. In this study, we tested serological and cellular response to SARS-CoV-2 mRNA vaccines in 16 patients treated with Ibrutinib, 16 treated with maintenance Rituximab, 18 patients with chronic lymphocytic leukaemia (CLL) with watch and wait status and 21 healthy volunteers. In comparison with the healthy volunteers, where serological response was achieved by 100% subjects, patients on B-cell-targeting therapy (Ibrutinib and Rituximab) had their response dramatically impaired. The serological response was achieved in 0% of Rituximab treated, 18% of Ibrutinib treated and 50% of untreated CLL patients. Cell-mediated immunity analysed by the whole blood Interferon-γ Release immune Assay developed in 80% of healthy controls, 62% of Rituximab treated, 75% of Ibrutinib treated and 55% of untreated CLL patients. The probability of cell-mediated immune response development negatively correlates with disease burden mainly in CLL patients. Our study shows that even though the serological response to SARS-CoV-2 vaccine is severely impaired in patients treated with B-cell-targeting therapy, the majority of these patients develop sufficient cell-mediated immunity. The vaccination of these patients therefore might be meaningful in terms of protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Barbora Bacova
- Department of Haematology, 3RdFaculty of Medicine, Faculty Hospital Kralovske Vinohrady, Charles University, Srobarova 50, 100 34, Prague 10, Czech Republic
- Central Laboratories of the Faculty Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Zuzana Kohutova
- Department of Haematology, 3RdFaculty of Medicine, Faculty Hospital Kralovske Vinohrady, Charles University, Srobarova 50, 100 34, Prague 10, Czech Republic
| | - Ivana Zubata
- Department of Haematology, 3RdFaculty of Medicine, Faculty Hospital Kralovske Vinohrady, Charles University, Srobarova 50, 100 34, Prague 10, Czech Republic
| | - Lubica Gaherova
- Department of Haematology, 3RdFaculty of Medicine, Faculty Hospital Kralovske Vinohrady, Charles University, Srobarova 50, 100 34, Prague 10, Czech Republic
| | - Petr Kucera
- Department of Immunology and Clinical Biochemistry, 3Rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomas Heizer
- Central Laboratories of the Faculty Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Marcela Mikesova
- Central Laboratories of the Faculty Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Tomas Karel
- Department of Statistics and Probability, Faculty of Informatics and Statistics, University of Economics and Business in Prague. Namesti W. Churchilla, 1938/4, 130 67, Prague 3, Czech Republic
| | - Jan Novak
- Department of Haematology, 3RdFaculty of Medicine, Faculty Hospital Kralovske Vinohrady, Charles University, Srobarova 50, 100 34, Prague 10, Czech Republic.
- Department of Immunology and Clinical Biochemistry, 3Rd Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
26
|
Töllner M, Speer C, Benning L, Bartenschlager M, Nusshag C, Morath C, Zeier M, Süsal C, Schnitzler P, Schmitt W, Bergner R, Bartenschlager R, Lorenz HM, Schaier M. Impaired Neutralizing Antibody Activity against B.1.617.2 (Delta) after Anti-SARS-CoV-2 Vaccination in Patients Receiving Anti-CD20 Therapy. J Clin Med 2022; 11:jcm11061739. [PMID: 35330069 PMCID: PMC8952324 DOI: 10.3390/jcm11061739] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 12/19/2022] Open
Abstract
Background: To characterize humoral response after standard anti-SARS-CoV-2 vaccination in Rituximab-treated patients and to determine the optimal time point after last Rituximab treatment for appropriate immunization. Methods: Sixty-four patients who received Rituximab within the last seven years prior to the first anti-SARS-CoV-2 vaccination were recruited in a prospective observational study. Anti-S1 IgG, SARS-CoV-2 specific neutralization, and various SARS-CoV-2 target antibodies were determined. A live virus assay was used to assess neutralizing antibody activity against B.1.617.2 (delta). In Rituximab-treated patients, CD19+ peripheral B-cells were quantified using flow cytometry. Results: After second vaccination, all antibodies were significantly reduced compared to healthy controls. Neutralizing antibody activity against B.1.617.2 (delta) was detectable with a median (IQR) ID50 of 0 (0−1:20) compared to 1:320 (1:160−1:320) in healthy controls (for all p < 0.001). Longer time period since last Rituximab administration correlated with higher anti-SARS-CoV-2 antibody levels and a stronger neutralization of B.1.617.2 (delta). With one exception, only patients with a CD19+ cell proportion ≥ 1% had detectable neutralizing antibodies. Conclusion: Our data indicate that a reconstitution of the B-cell population to >1% seems crucial in developing neutralizing antibodies against SARS-CoV-2. We suggest that anti-SARS-CoV-2 vaccination should be administered at least 8−12 months after the last Rituximab treatment for sufficient humoral responses.
Collapse
Affiliation(s)
- Maximilian Töllner
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
- Correspondence:
| | - Claudius Speer
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
- Molecular Medicine Partnership Unit Heidelberg, European Molecular Biology Laboratory, 69120 Heidelberg, Germany
| | - Louise Benning
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| | - Marie Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, 69120 Heidelberg, Germany; (M.B.); (R.B.)
| | - Christian Nusshag
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| | - Christian Morath
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| | - Caner Süsal
- Transplant Immunology Research Center of Excellence, Koç University Hospital, Istanbul 34010, Turkey;
| | - Paul Schnitzler
- Department of Infectious Diseases, Virology, University of Heidelberg, 69120 Heidelberg, Germany;
| | | | - Raoul Bergner
- Clinical Center Ludwigshafen, Department of Internal Medicine A, 67036 Ludwigshafen, Germany;
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, 69120 Heidelberg, Germany; (M.B.); (R.B.)
- German Center for Infection Research (DZIF), Heidelberg Partner Site, 69120 Heidelberg, Germany
- Division Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Department of Medicine V, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Matthias Schaier
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany; (C.S.); (L.B.); (C.N.); (C.M.); (M.Z.); (M.S.)
| |
Collapse
|
27
|
Dalla Costa G, Leocani L, Comi G. Ofatumumab subcutaneous injection for the treatment of relapsing forms of multiple sclerosis. Expert Rev Clin Immunol 2022; 18:105-114. [PMID: 35107057 DOI: 10.1080/1744666x.2022.2031982] [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: 11/04/2022]
Abstract
INTRODUCTION In recent years, different studies have highlighted the importance of B cells in the pathophysiology of multiple sclerosis (MS): they secrete cytokines to modulate the inflammatory environment, present antigens for the activation of T lymphocytes, and they secrete antibodies contributing to the destruction of the myelin sheath. Combined, these findings have lead to new possible means for treating MS. AREAS COVERED In this review, we provide an up-to-date overview of the characteristics of ofatumumab (aka Kesimpta), and the differences between this drug and the other anti-CD20 monoclonal antibodies used to treat MS. EXPERT OPINION The evolution of disease-modifying treatment algorithms in MS underlines the importance of starting treatment as soon as the diagnosis is defined, and with adequate "treatment intensity". Monoclonal antibodies and other aggressive treatments are now considered as an option at the clinical presentation of the disease, based to the prognostic profile emerging through clinical and paraclinical investigations. The recent adoption of new diagnostic criteria allows for the early diagnosis of MS. This, together with the availability of disease-modifying therapies (DMTs), such as ofatumumab, with a good efficacy/safety profile and which are easy to administer, could contribute to significant improvements in the long-term prognosis of MS.
Collapse
Affiliation(s)
- G Dalla Costa
- Institute of Experimental Neurophysiology of San Raffaele Hospital, via Olgettina 60, Milan, Italy.,Vita-Salute San Raffaele University, via Olgettina 60, Milan, Italy
| | - L Leocani
- Institute of Experimental Neurophysiology of San Raffaele Hospital, via Olgettina 60, Milan, Italy.,Vita-Salute San Raffaele University, via Olgettina 60, Milan, Italy
| | - G Comi
- Vita-Salute San Raffaele University, via Olgettina 60, Milan, Italy.,Casa di Cura Privata del Policlinico, via Dezza 48, Milan, Italy
| |
Collapse
|
28
|
Galmiche S, Luong Nguyen LB, Tartour E, de Lamballerie X, Wittkop L, Loubet P, Launay O. Immunological and clinical efficacy of COVID-19 vaccines in immunocompromised populations: a systematic review. Clin Microbiol Infect 2022; 28:163-177. [PMID: 35020589 PMCID: PMC8595936 DOI: 10.1016/j.cmi.2021.09.036] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND Available data show that COVID-19 vaccines may be less effective in immunocompromised populations, who are at increased risk of severe COVID-19. OBJECTIVES We conducted a systematic review of literature to assess immunogenicity, efficacy and effectiveness of COVID-19 vaccines in immunocompromised populations. DATA SOURCES We searched Medline and Embase databases. STUDY ELIGIBILITY CRITERIA, PATIENTS, INTERVENTIONS We included studies of COVID-19 vaccines after complete vaccination in immunocompromised patients until 31 August 2021. Studies with <10 patients, safety data only and case series of breakthrough infections were excluded. METHODS Risk of bias was assessed via the tool developed by the National Institutes of Health on interventional and observational studies. Immunogenicity was assessed through non-response rate defined as no anti-SARS-CoV-2 spike protein antibodies, efficacy and effectiveness by the relative reduction in risk of SARS-CoV-2 infection or COVID-19. We collected factors associated with the risk of non-response. We presented collected data by immunosuppression type. RESULTS We screened 5917 results, included 162 studies. There were 157 on immunogenicity in 25 209 participants, including 7835 cancer or haematological malignancy patients (31.1%), 6302 patients on dialysis (25.0%), 5974 solid organ transplant recipients (23.7%) and 4680 immune-mediated disease patients (18.6%). Proportion of non-responders seemed higher among solid organ transplant recipients (range 18-100%) and patients with haematological malignancy (range 14-61%), and lower in patients with cancer (range 2-36%) and patients on dialysis (range 2-30%). Risk factors for non-response included older age, use of corticosteroids, immunosuppressive or anti-CD20 agent. Ten studies evaluated immunogenicity of an additional dose. Five studies evaluated vaccine efficacy or effectiveness: three on SARS-CoV-2 infection (range 71-81%), one on COVID-19-related hospitalization (62.9%), one had a too small sample size. CONCLUSIONS This systematic review highlights the risk of low immunogenicity of COVID-19 vaccines in immunocompromised populations, especially solid organ transplant recipients and patients with haematological malignancy. Despite lack of vaccine effectiveness data, enhanced vaccine regimens may be necessary.
Collapse
Affiliation(s)
- Simon Galmiche
- Assistance Publique - Hôpitaux de Paris (AP-HP), CIC Cochin Pasteur, Hôpital Cochin, Paris, France
| | - Liem Binh Luong Nguyen
- Assistance Publique - Hôpitaux de Paris (AP-HP), CIC Cochin Pasteur, Hôpital Cochin, Paris, France
| | - Eric Tartour
- AP-HP, Immunologie Biologique, Hôpital Européen Georges Pompidou, Paris, France
| | - Xavier de Lamballerie
- Aix Marseille Université, IRD 190, INSERM 1207, Unité des Virus Emergents, UVE, IHU Méditerranée Infection, Marseille, France
| | - Linda Wittkop
- Institut de Santé Publique d'Epidémiologie et de Développement, INSERM, Bordeaux Population Health Research Center, UMR 1219, Centre d'Investigation Clinique-Epidémiologie Clinique 1401, University of Bordeaux, Service d'Information Médicale, CHU de Bordeaux, Pôle de Santé Publique, Bordeaux, France
| | - Paul Loubet
- INSERM U1047, Department of Infectious and Tropical Diseases, CHU Nîmes, Université Montpellier, Nîmes, France
| | - Odile Launay
- Université de Paris, Faculté de Médecine Paris Descartes, AP-PH, Inserm, CIC Cochin Pasteur, Paris, France.
| |
Collapse
|
29
|
Schietzel S, Anderegg M, Limacher A, Born A, Horn MP, Maurer B, Hirzel C, Sidler D, Moor MB. Humoral and cellular immune responses on SARS-CoV-2 vaccines in patients with anti-CD20 therapies: a systematic review and meta-analysis of 1342 patients. RMD Open 2022; 8:e002036. [PMID: 35115385 PMCID: PMC8814433 DOI: 10.1136/rmdopen-2021-002036] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Immune responses on SARS-CoV-2 vaccination in patients receiving anti-CD20 therapies are impaired but vary considerably. We conducted a systematic review and meta-analysis of the literature on SARS-CoV-2 vaccine induced humoral and cell-mediated immune response in patients previously treated with anti-CD20 antibodies. METHODS We searched PubMed, Embase, Medrxiv and SSRN using variations of search terms 'anti-CD20', 'vaccine' and 'COVID' and included original studies up to 21 August 2021. We excluded studies with missing data on humoral or cell-mediated immune response, unspecified methodology of response testing, unspecified timeframes between vaccination and blood sampling or low number of participants (≤3). We excluded individual patients with prior COVID-19 or incomplete vaccine courses. Primary endpoints were humoral and cell-mediated immune response rates. Subgroup analyses included time since anti-CD20 therapy, B cell depletion and indication for anti-CD20 therapy. We used random-effects models of proportions. FINDINGS Ninety studies were assessed. Inclusion criteria were met by 23 studies comprising 1342 patients. Overall rate of humoral response was 0.40 (95% CI 0.35 to 0.47). Overall rate of cell-mediated immune responses was 0.71 (95% CI 0.57 to 0.87). A time interval >6 months since last anti-CD20 therapy was associated with higher humoral response rates with 0.63 (95% CI 0.53 to 0.72) versus <6 months 0.2 (95% CI 0.03 to 0.43); p=0<01. Similarly, patients with circulating B cells more frequently showed humoral responses. Anti-CD20-treated kidney transplant recipients showed lower humoral response rates than patients with haematological malignancies or autoimmune disease. INTERPRETATION Patients on anti-CD20 therapies can develop humoral and cell-mediated immune responses after SARS-CoV-2 vaccination, but subgroups such as kidney transplant recipients or those with very recent therapy and depleted B cell are at high risk for non-seroconversion and should be individually assessed for personalised SARS-CoV-2 vaccination strategies. Potential limitations are small patient numbers and heterogeneity of studies included. FUNDING This study was funded by Bern University Hospital.
Collapse
Affiliation(s)
- Simeon Schietzel
- Department of Nephrology and Hypertension, Inselspital University Hospital Bern, Bern, Switzerland
| | - Manuel Anderegg
- Department of Nephrology and Hypertension, Inselspital University Hospital Bern, Bern, Switzerland
- Division of Nephrology, Department of Internal Medicine, Neuchâtel Hospital Network, Neuchâtel, Switzerland
| | | | - Alexander Born
- Department of Nephrology and Hypertension, Inselspital University Hospital Bern, Bern, Switzerland
| | - Michael P Horn
- Department of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Britta Maurer
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Cedric Hirzel
- Department of Infectious Diseases, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Daniel Sidler
- Department of Nephrology and Hypertension, Inselspital University Hospital Bern, Bern, Switzerland
| | - Matthias B Moor
- Department of Nephrology and Hypertension, Inselspital University Hospital Bern, Bern, Switzerland
| |
Collapse
|
30
|
Hagihara M, Imai Y, Uchida T, Ohara S, Inoue M, Sugi T, Mitamura K. Successful Elimination of SARS-CoV-2 Following Vaccination with BNT162b2 after Prolonged Viral Infection in an Immunocompromised Lymphoma Patient. Intern Med 2022; 61:2215-2219. [PMID: 35850990 PMCID: PMC9381351 DOI: 10.2169/internalmedicine.9513-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A 52-year-old man with mantle cell lymphoma treated with bendamustine and rituximab developed prolonged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Despite elevated titers of anti-spike IgG antibody, protracted pancytopenia persisted for more than six months. Finally, the anti-SARS CoV-2 vaccine, BNT162b2, was administered, which improved his blood cell count and eliminated the virus. The increased anti-spike IgG titer and lymphocyte count after vaccination suggested that both humoral and cellular immunity acted in coordination to eliminate the virus.
Collapse
Affiliation(s)
| | - Yui Imai
- Department of Hematology, Eiju General Hospital, Japan
| | | | - Shin Ohara
- Department of Hematology, Eiju General Hospital, Japan
| | | | - Tomiyuki Sugi
- Department of Pharmacy, Eiju General Hospital, Japan
| | - Keiko Mitamura
- Department of Preventive Medicine, Eiju General Hospital, Japan
| |
Collapse
|
31
|
Ercoli G, Ramos‐Sevillano E, Pearce E, Ragab S, Goldblatt D, Weckbecker G, Brown JS. Maintained partial protection against Streptococcus pneumoniae despite B-cell depletion in mice vaccinated with a pneumococcal glycoconjugate vaccine. Clin Transl Immunology 2022; 11:e1366. [PMID: 35003749 PMCID: PMC8715227 DOI: 10.1002/cti2.1366] [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: 06/08/2021] [Revised: 11/16/2021] [Accepted: 12/16/2021] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES Anti-CD20 monoclonal antibody therapy rapidly depletes > 95% of CD20+ B cells from the circulation. B-cell depletion is an effective treatment for autoimmune disease and B-cell malignancies but also increases the risk of respiratory tract infections. This effect on adaptive immunity could be countered by vaccination. We have used mouse models to investigate the effects of B-cell depletion on pneumococcal vaccination, including protection against infection and timing of vaccination in relation to B-cell depletion. METHODS C57BL/6 female mice were B-cell depleted using anti-CD20 antibody and immunized with two doses of Prevnar-13 vaccine either before or after anti-CD20 treatment. B-cell repertoire and Streptococcus pneumoniae-specific IgG levels were measured using whole-cell ELISA and flow cytometry antibody-binding assay. Protection induced by vaccination was assessed by challenging the mice using a S. pneumoniae pneumonia model. RESULTS Antibody responses to S. pneumoniae were largely preserved in mice B-cell depleted after vaccination resulting in full protection against pneumococcal infections. In contrast, mice vaccinated with Prevnar-13 while B cells were depleted (with > 90% reduction in B-cell numbers) had decreased circulating anti-S. pneumoniae IgG and IgM levels (measured using ELISA and flow cytometry antibody binding assays). However, some antibody responses were maintained, and, although vaccine-induced protection against S. pneumoniae infection was impaired, septicaemia was still prevented in 50% of challenged mice. CONCLUSIONS This study showed that although vaccine efficacy during periods of profound B-cell depletion was impaired some protective efficacy was preserved, suggesting that vaccination remains beneficial.
Collapse
Affiliation(s)
- Giuseppe Ercoli
- Centre for Inflammation and Tissue RepairUCL RespiratoryDivision of MedicineUniversity College Medical SchoolRayne InstituteLondonUK
| | - Elisa Ramos‐Sevillano
- Centre for Inflammation and Tissue RepairUCL RespiratoryDivision of MedicineUniversity College Medical SchoolRayne InstituteLondonUK
| | - Emma Pearce
- Department of ImmunobiologyUCL Great Ormond Street Institute of Child HealthNIHR Biomedical Research CentreLondonUK
| | - Sara Ragab
- Department of ImmunobiologyUCL Great Ormond Street Institute of Child HealthNIHR Biomedical Research CentreLondonUK
| | - David Goldblatt
- Department of ImmunobiologyUCL Great Ormond Street Institute of Child HealthNIHR Biomedical Research CentreLondonUK
| | | | - Jeremy S Brown
- Centre for Inflammation and Tissue RepairUCL RespiratoryDivision of MedicineUniversity College Medical SchoolRayne InstituteLondonUK
| |
Collapse
|
32
|
Law MF, Ho R, Law KWT, Cheung CKM. Gastrointestinal and hepatic side effects of potential treatment for COVID-19 and vaccination in patients with chronic liver diseases. World J Hepatol 2021; 13:1850-1874. [PMID: 35069994 PMCID: PMC8727202 DOI: 10.4254/wjh.v13.i12.1850] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/20/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) is a global pandemic. Many clinical trials have been performed to investigate potential treatments or vaccines for this disease to reduce the high morbidity and mortality. The drugs of higher interest include umifenovir, bromhexine, remdesivir, lopinavir/ritonavir, steroid, tocilizumab, interferon alpha or beta, ribavirin, fivapiravir, nitazoxanide, ivermectin, molnupiravir, hydroxychloroquine/chloroquine alone or in combination with azithromycin, and baricitinib. Gastrointestinal (GI) symptoms and liver dysfunction are frequently seen in patients with COVID-19, which can make it difficult to differentiate disease manifestations from treatment adverse effects. GI symptoms of COVID-19 include anorexia, dyspepsia, nausea, vomiting, diarrhea and abdominal pain. Liver injury can be a result of systemic inflammation or cytokine storm, or due to the adverse drug effects in patients who have been receiving different treatments. Regular monitoring of liver function should be performed. COVID-19 vaccines have been rapidly developed with different technologies including mRNA, viral vectors, inactivated viruses, recombinant DNA, protein subunits and live attenuated viruses. Patients with chronic liver disease or inflammatory bowel disease and liver transplant recipients are encouraged to receive vaccination as the benefits outweigh the risks. Vaccination against COVID-19 is also recommended to family members and healthcare professionals caring for these patients to reduce exposure to the severe acute respiratory syndrome coronavirus 2 virus.
Collapse
Affiliation(s)
- Man Fai Law
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
| | - Rita Ho
- Department of Medicine, North District Hospital, Hong Kong, China
| | | | - Carmen Ka Man Cheung
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
33
|
Shields AM, Venkatachalam S, Shafeek S, Paneesha S, Ford M, Sheeran T, Kelly M, Qureshi I, Salhan B, Karim F, De Silva N, Stones J, Lee S, Khawaja J, Kaudlay PK, Whitmill R, Kakepoto GN, Parry HM, Moss P, Faustini SE, Richter AG, Drayson MT, Basu S. SARS-CoV-2 vaccine responses following CD20-depletion treatment in patients with haematological and rheumatological disease: a West Midlands Research Consortium study. Clin Exp Immunol 2021; 207:3-10. [PMID: 35020852 PMCID: PMC8767851 DOI: 10.1093/cei/uxab018] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022] Open
Abstract
B-cell-depleting agents are among the most commonly used drugs to treat haemato-oncological and autoimmune diseases. They rapidly induce a state of peripheral B-cell aplasia with the potential to interfere with nascent vaccine responses, particularly to novel antigens. We have examined the relationship between B-cell reconstitution and SARS-CoV-2 vaccine responses in two cohorts of patients previously exposed to B-cell-depleting agents: a cohort of patients treated for haematological B-cell malignancy and another treated for rheumatological disease. B-cell depletion severely impairs vaccine responsiveness in the first 6 months after administration: SARS-CoV-2 antibody seroprevalence was 42.2% and 33.3% in the haemato-oncological patients and rheumatology patients, respectively and 22.7% in patients vaccinated while actively receiving anti-lymphoma chemotherapy. After the first 6 months, vaccine responsiveness significantly improved during early B-cell reconstitution; however, the kinetics of reconstitution was significantly faster in haemato-oncology patients. The AstraZeneca ChAdOx1 nCoV-19 vaccine and the Pfizer BioNTech 162b vaccine induced equivalent vaccine responses; however, shorter intervals between vaccine doses (<1 m) improved the magnitude of the antibody response in haeamto-oncology patients. In a subgroup of haemato-oncology patients, with historic exposure to B-cell-depleting agents (>36 m previously), vaccine non-responsiveness was independent of peripheral B-cell reconstitution. The findings have important implications for primary vaccination and booster vaccination strategies in individuals clinically vulnerable to SARS-CoV-2.
Collapse
Affiliation(s)
- Adrian M Shields
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Department of Clinical Immunology, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK,Correspondence: Adrian M. Shields, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| | | | - Salim Shafeek
- Department of Haematology, Worcestershire Acute NHS Trust, Worcester, UK
| | - Shankara Paneesha
- Department of Haematology, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - Mark Ford
- Department of Rheumatology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Tom Sheeran
- Department of Rheumatology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Melanie Kelly
- Department of Haematology, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - Iman Qureshi
- Department of Haematology, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - Beena Salhan
- Department of Haematology, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - Farheen Karim
- Department of Haematology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Neelakshi De Silva
- Department of Haematology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Jacqueline Stones
- Department of Haematology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Sophie Lee
- Department of Haematology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Jahanzeb Khawaja
- Department of Haematology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | | | - Richard Whitmill
- Department of Haematology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | | | - Helen M Parry
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Department of Haematology, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Department of Haematology, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - Sian E Faustini
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Alex G Richter
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Department of Clinical Immunology, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - Mark T Drayson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Department of Clinical Immunology, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK,Mark T. Drayson, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| | - Supratik Basu
- Department of Haematology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK,Faculty of Science & Engineering, University of Wolverhampton, Wolverhampton, UK,Supratik Basu, The Royal Wolverhampton NHS Trust, Wolverhampton, UK.
| |
Collapse
|
34
|
Magliulo D, Wade SD, Kyttaris VC. Immunogenicity of SARS-CoV-2 vaccination in rituximab-treated patients: Effect of timing and immunologic parameters. Clin Immunol 2021; 234:108897. [PMID: 34848357 PMCID: PMC8627008 DOI: 10.1016/j.clim.2021.108897] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022]
Abstract
Rituximab (RTX), an important therapeutic option for patients with rheumatic diseases, has been shown to reduce immune responses to various vaccines. We asked whether following SARS-CoV-2 vaccination, response rates in RTX treated patients are reduced and whether specific patient characteristics influence the responses. We recruited patients on chronic RTX therapy undergoing anti-SARS-CoV2 vaccination and measured the post-vaccination anti-spike IgG antibody levels. The median time from pre-vaccination RTX infusion to vaccination and from vaccination to the post-vaccination RTX infusion was 20.5 weeks and 7.2 weeks respectively. Only 36.5% of patients developed measurable titers of IgG anti-SARS-CoV-2 spike antibody after vaccination. Hypogammaglobulinemia (IgG and/or IgM) but not timing of vaccination, B cell numbers, or concomitant immune suppressive medications, correlated with sero-negativity (p = 0.004). Our results underscore the fact that even after B cell reconstitution, RTX induced chronic hypogammaglobulinemia significantly impairs the ability of the immune system to respond to SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Daniel Magliulo
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - Stefanie D Wade
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - Vasileios C Kyttaris
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
35
|
Becker J, Ferreira LC, Damasceno A, Bichuetti DB, Christo PP, Callegaro D, Peixoto MAL, Sousa NADC, Almeida SMD, Adoni T, Santiago-Amaral J, Junqueira T, Pereira SLA, Gomes ABAGR, Pitombeira M, Paolilo RB, Grzesiuk AK, Piccolo AC, D Almeida JAC, Gomes Neto AP, Oliveira ACPD, Oliveira BSD, Tauil CB, Vasconcelos CF, Kaimen-Maciel D, Varela D, Diniz DS, Oliveira EMLD, Malfetano FR, Borges FE, Figueira FFA, Gondim FDAA, Passos GRD, Silva GD, Olival GSD, Santos GACD, Ruocco HH, Sato HK, Soares Neto HR, Cortoni Calia L, Gonçalves MVM, Vecino MCAD, Pimentel MLV, Ribeiro MDC, Boaventura M, Parolin MKF, Melo RBDS, Lázaro R, Thomaz RB, Kleinpaul R, Dias RM, Gomes S, Lucatto SA, Alves-Leon SV, Fukuda T, Ribeiro TAGJ, Winckler TCD, Fragoso YD, Nascimento OJMD, Ferreira MLB, Mendes MF, Brum DG, Glehn FV. Recommendations by the Scientific Department of Neuroimmunology of the Brazilian Academy of Neurology (DCNI/ABN) and the Brazilian Committee for Treatment and Research in Multiple Sclerosis and Neuroimmunological Diseases (BCTRIMS) on vaccination in general and specifically against SARS-CoV-2 for patients with demyelinating diseases of the central nervous system. ARQUIVOS DE NEURO-PSIQUIATRIA 2021; 79:1049-1061. [PMID: 34816999 DOI: 10.1590/0004-282x-anp-2021-0162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 05/30/2021] [Indexed: 11/22/2022]
Abstract
The Scientific Department of Neuroimmunology of the Brazilian Academy of Neurology (DCNI/ABN) and Brazilian Committee for Treatment and Research in Multiple Sclerosis and Neuroimmunological Diseases (BCTRIMS) provide recommendations in this document for vaccination of the population with demyelinating diseases of the central nervous system (CNS) against infections in general and against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes COVID-19. We emphasize the seriousness of the current situation in view of the spread of COVID-19 in our country. Therefore, reference guides on vaccination for clinicians, patients, and public health authorities are particularly important to prevent some infectious diseases. The DCNI/ABN and BCTRIMS recommend that patients with CNS demyelinating diseases (e.g., MS and NMOSD) be continually monitored for updates to their vaccination schedule, especially at the beginning or before a change in treatment with a disease modifying drug (DMD). It is also important to note that vaccines are safe, and physicians should encourage their use in all patients. Clearly, special care should be taken when live attenuated viruses are involved. Finally, it is important for physicians to verify which DMD the patient is receiving and when the last dose was taken, as each drug may affect the induction of immune response differently.
Collapse
Affiliation(s)
- Jefferson Becker
- Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre RS, Brazil
| | - Lis Campos Ferreira
- Universidade Federal de Sergipe, Aracaju SE, Brazil.,Universidade Tiradentes, Aracaju SE, Brazil
| | - Alfredo Damasceno
- Universidade de Campinas, Faculdade de Ciências Médicas, Campinas SP, Brazil
| | | | | | - Dagoberto Callegaro
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, São Paulo SP, Brazil
| | | | | | | | - Tarso Adoni
- Hospital Sírio Libanês, São Paulo SP, Brazil
| | | | | | | | | | | | - Renata Barbosa Paolilo
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, São Paulo SP, Brazil
| | | | | | | | | | | | | | | | | | | | - Daniel Varela
- Hospital de Clínicas de Passo Fundo, Passo Fundo RS, Brazil
| | | | | | | | | | | | | | | | - Guilherme Diogo Silva
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, São Paulo SP, Brazil
| | | | | | - Heloisa Helena Ruocco
- Universidade Federal Fluminense, Niterói RJ, Brazil.,Pontifícia Universidade Católica, Campina SP, Brazil
| | | | | | | | | | | | | | | | - Mateus Boaventura
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, São Paulo SP, Brazil
| | | | | | - Robson Lázaro
- Faculdade de Medicina de Jundiaí, Jundiaí SP, Brazil
| | | | | | | | - Sidney Gomes
- Hospital Beneficiência Portuguesa, São Paulo SP, Brazil
| | | | | | - Thiago Fukuda
- Hospital Universitário Prof. Edgar Santos, Salvador BA, Brazil
| | | | | | | | | | | | | | | | - Felipe Von Glehn
- Universidade de Campinas, Faculdade de Ciências Médicas, Campinas SP, Brazil.,Universidade de Brasília, Faculdade de Medicina, Brasília DF, Brazil
| |
Collapse
|
36
|
Beard K, Sriwastava S. Insight in booster COVID-19 vaccine and disease modifying therapy in multiple sclerosis. J Neurol Sci 2021; 430:120034. [PMID: 34688988 PMCID: PMC8523307 DOI: 10.1016/j.jns.2021.120034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/07/2021] [Accepted: 10/15/2021] [Indexed: 01/04/2023]
Affiliation(s)
- Katherine Beard
- School of Medicine, West Virginia University, United States of America,Department of Neurology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States of America
| | - Shitiz Sriwastava
- School of Medicine, West Virginia University, United States of America,Department of Neurology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States of America,West Virginia Clinical and Translational Science Institute, United States of America,Corresponding author at: Department of Neurology, Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506, United States of America
| |
Collapse
|
37
|
Haskin O, Ashkenazi-Hoffnung L, Ziv N, Borovitz Y, Dagan A, Levi S, Koren G, Hamdani G, Levi-Erez D, Landau D, Alfandary H. Serological Response to the BNT162b2 COVID-19 mRNA Vaccine in Adolescent and Young Adult Kidney Transplant Recipients. Transplantation 2021; 105:e226-e233. [PMID: 34381004 PMCID: PMC8549126 DOI: 10.1097/tp.0000000000003922] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/21/2021] [Accepted: 07/28/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Initial reports in adult kidney transplant recipients (KTR) indicate low immunogenicity after 2 doses of the BNT162b2 COVID-19 mRNA vaccine. We describe the immunogenicity of this vaccine compared to the serologic response in naturally infected COVID-19 positive adolescent and young adult KTR. METHODS For this prospective observational study, the study group included 38 KTR who received 2 doses of the tested vaccine, and the control group included 14 KTR who had a previous polymerase chain reaction-confirmed COVID-19 infection. RESULTS The mean age was 18 ± 3 y. Positive serologic responses were observed in 63% and 100% of the study and control groups, respectively (P = 0.01). Antibody titers were almost 30-fold higher in the control than the study group (median [interquartile range (IQR)]: 2782 [1908-11 000] versus 100.3 [4.7-1744] AU/mL, P < 0.001), despite the longer time from the COVID-19 infection to serologic testing compared to time from vaccination (median [IQR]: 157.5 [60-216] versus 37 [20.5-53] d, P = 0.011). Among vaccinated patients, higher proportions of those seronegative than seropositive were previously treated with rituximab (50% versus 8%, P = 0.01). Time from the second vaccine dose to serologic testing was longer in seropositive than seronegative patients (median [IQR]: 24.5 [15-40] versus 46 [27-56] d, P = 0.05). No patient developed symptomatic COVID-19 disease postvaccination. CONCLUSIONS The BNT162b2 COVID-19 mRNA vaccine yielded higher positive antibody response in adolescent and young adult KTR than previously reported for adult KTR. Antibody titers after vaccination were significantly lower than following COVID-19 infection. Longer time may be required to mount appropriate humoral immunity to vaccination in KTR.
Collapse
Affiliation(s)
- Orly Haskin
- Institute of Nephrology, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Liat Ashkenazi-Hoffnung
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Infectious Disease Unit, Day Hospitalization Department, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
| | - Noa Ziv
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Pediatrics “C”, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
| | - Yael Borovitz
- Institute of Nephrology, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
| | - Amit Dagan
- Institute of Nephrology, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shelly Levi
- Institute of Nephrology, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gili Koren
- Institute of Nephrology, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
| | - Gilad Hamdani
- Institute of Nephrology, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
| | - Daniella Levi-Erez
- Institute of Nephrology, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daniel Landau
- Institute of Nephrology, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hadas Alfandary
- Institute of Nephrology, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
38
|
Apostolidis SA, Kakara M, Painter MM, Goel RR, Mathew D, Lenzi K, Rezk A, Patterson KR, Espinoza DA, Kadri JC, Markowitz DM, E Markowitz C, Mexhitaj I, Jacobs D, Babb A, Betts MR, Prak ETL, Weiskopf D, Grifoni A, Lundgreen KA, Gouma S, Sette A, Bates P, Hensley SE, Greenplate AR, Wherry EJ, Li R, Bar-Or A. Cellular and humoral immune responses following SARS-CoV-2 mRNA vaccination in patients with multiple sclerosis on anti-CD20 therapy. Nat Med 2021; 27:1990-2001. [PMID: 34522051 PMCID: PMC8604727 DOI: 10.1038/s41591-021-01507-2] [Citation(s) in RCA: 365] [Impact Index Per Article: 91.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/16/2021] [Indexed: 02/08/2023]
Abstract
SARS-CoV-2 messenger RNA vaccination in healthy individuals generates immune protection against COVID-19. However, little is known about SARS-CoV-2 mRNA vaccine-induced responses in immunosuppressed patients. We investigated induction of antigen-specific antibody, B cell and T cell responses longitudinally in patients with multiple sclerosis (MS) on anti-CD20 antibody monotherapy (n = 20) compared with healthy controls (n = 10) after BNT162b2 or mRNA-1273 mRNA vaccination. Treatment with anti-CD20 monoclonal antibody (aCD20) significantly reduced spike-specific and receptor-binding domain (RBD)-specific antibody and memory B cell responses in most patients, an effect ameliorated with longer duration from last aCD20 treatment and extent of B cell reconstitution. By contrast, all patients with MS treated with aCD20 generated antigen-specific CD4 and CD8 T cell responses after vaccination. Treatment with aCD20 skewed responses, compromising circulating follicular helper T (TFH) cell responses and augmenting CD8 T cell induction, while preserving type 1 helper T (TH1) cell priming. Patients with MS treated with aCD20 lacking anti-RBD IgG had the most severe defect in circulating TFH responses and more robust CD8 T cell responses. These data define the nature of the SARS-CoV-2 vaccine-induced immune landscape in aCD20-treated patients and provide insights into coordinated mRNA vaccine-induced immune responses in humans. Our findings have implications for clinical decision-making and public health policy for immunosuppressed patients including those treated with aCD20.
Collapse
Grants
- U19AI082630 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- T32 AR076951 NIAMS NIH HHS
- AI082630 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- R21 AI142638 NIAID NIH HHS
- AI108545 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- R01 AI152236 NIAID NIH HHS
- 75N9301900065 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- AI149680 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- T32 CA009140 NCI NIH HHS
- R01 AI118694 NIAID NIH HHS
- U19 AI082630 NIAID NIH HHS
- AI152236 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- P30-AI0450080 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- T32 AR076951-01 U.S. Department of Health & Human Services | NIH | National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
- R01 AI105343 NIAID NIH HHS
- AI105343 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- R01 AI155577 NIAID NIH HHS
- UM1 AI144288 NIAID NIH HHS
- U19 AI149680 NIAID NIH HHS
- AI155577 U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER)
- SI-2011-37160 National Multiple Sclerosis Society (National MS Society)
- UC4 DK112217 NIDDK NIH HHS
- P01 AI108545 NIAID NIH HHS
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases (Division of Intramural Research of the NIAID)
- Penn | Perelman School of Medicine, University of Pennsylvania (Perelman School of Medicine at the University of Pennsylvania)
Collapse
Affiliation(s)
- Sokratis A Apostolidis
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mihir Kakara
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mark M Painter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rishi R Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Divij Mathew
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kerry Lenzi
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ayman Rezk
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kristina R Patterson
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Diego A Espinoza
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immunology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessy C Kadri
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel M Markowitz
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Clyde E Markowitz
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ina Mexhitaj
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Dina Jacobs
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison Babb
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael R Betts
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Eline T Luning Prak
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Kendall A Lundgreen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Penn Center for Research on Coronavirus and Other Emerging Pathogens, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sigrid Gouma
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, CA, USA
| | - Paul Bates
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Penn Center for Research on Coronavirus and Other Emerging Pathogens, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott E Hensley
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison R Greenplate
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - E John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Rui Li
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
39
|
MENACTRIMS practice guideline for COVID-19 vaccination in patients with multiple sclerosis. Mult Scler Relat Disord 2021; 56:103225. [PMID: 34479111 PMCID: PMC8386106 DOI: 10.1016/j.msard.2021.103225] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 12/30/2022]
Abstract
Patients with multiple sclerosis (MS) should be vaccinated against COVID-19. All COVID-19 vaccines are effective and do not appear to carry any additional risk for patients with MS. Patients with MS should get a COVID-19 vaccine as soon as it becomes available. The risks of COVID-19 disease outweigh any potential risks from the vaccine. Even if vaccinated, patients with MS should continue to practice standard and recommended precautions against COVID-19, such as wearing a face mask, social distancing and washing hands. There is no evidence that patients with MS are at higher risk of complications from the mRNA, non-replicating viral vector, inactivated virus or protein COVID-19 vaccines, compared to the general population. COVID-19 Vaccines are safe to use in patients with MS treated with disease-modifying therapies (DMTs). The effectiveness of vaccination may be affected by few of the DMTs but yet some protection is still provided. For certain DMTs we may consider coordinating the timing of the vaccine with the timing of the DMT dose to increase vaccine efficacy.
Collapse
|
40
|
A prospective multicenter study assessing humoral immunogenicity and safety of the mRNA SARS-CoV-2 vaccines in Greek patients with systemic autoimmune and autoinflammatory rheumatic diseases. J Autoimmun 2021; 125:102743. [PMID: 34757289 PMCID: PMC8552665 DOI: 10.1016/j.jaut.2021.102743] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/23/2021] [Accepted: 10/24/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES To investigate humoral responses and safety of mRNA SARS-CoV-2 vaccines in systemic autoimmune and autoinflammatory rheumatic disease (SAARD) patients subjected or not to treatment modifications during vaccination. METHODS A nationwide, multicenter study, including 605 SAARD patients and 116 controls, prospectively evaluated serum anti-SARS-CoV-2 S1-protein IgG antibody titers, side-effects, and disease activity, one month after complete vaccination, in terms of distinct treatment modification strategies (none, partial and extended modifications). Independent risk factors associated with hampered humoral responses were identified by data-driven multivariable logistic regression analysis. RESULTS Patients with extended treatment modifications responded to vaccines similarly to controls as well as SAARD patients without immunosuppressive therapy (97.56% vs 100%, p = 0.2468 and 97.56% vs 97.46%, p > 0.9999, respectively). In contrast, patients with partial or without therapeutic modifications responded in 87.50% and 84.50%, respectively. Furthermore, SAARD patients with extended treatment modifications developed higher anti-SARS-CoV-2 antibody levels compared to those without or with partial modifications (median:7.90 vs 7.06 vs 7.1, p = 0.0003 and p = 0.0195, respectively). Mycophenolate mofetil (MMF), rituximab (RTX) and methotrexate (MTX) negatively affected anti-SARS-CoV-2 humoral responses. In 10.5% of vaccinated patients, mild clinical deterioration was noted; however, no differences in the incidence of deterioration were observed among the distinct treatment modification SAARD subgroups. Side-effects were generally comparable between SAARD patients and controls. CONCLUSIONS In SAARD patients, mRNA SARS-CoV-2 vaccines are effective and safe, both in terms of side-effects and disease flares. Treatment with MMF, RTX and/or MTX compromises anti-SARS-CoV-2 antibody responses, which are restored upon extended treatment modifications without affecting disease activity.
Collapse
|
41
|
Furer V, Eviatar T, Zisman D, Peleg H, Paran D, Levartovsky D, Zisapel M, Elalouf O, Kaufman I, Meidan R, Broyde A, Polachek A, Wollman J, Litinsky I, Meridor K, Nochomovitz H, Silberman A, Rosenberg D, Feld J, Haddad A, Gazzit T, Elias M, Higazi N, Kharouf F, Shefer G, Sharon O, Pel S, Nevo S, Elkayam O. Immunogenicity and safety of the BNT162b2 mRNA COVID-19 vaccine in adult patients with autoimmune inflammatory rheumatic diseases and in the general population: a multicentre study. Ann Rheum Dis 2021; 80:1330-1338. [PMID: 34127481 PMCID: PMC8206170 DOI: 10.1136/annrheumdis-2021-220647] [Citation(s) in RCA: 470] [Impact Index Per Article: 117.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/27/2021] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Vaccination represents a cornerstone in mastering the COVID-19 pandemic. Data on immunogenicity and safety of messenger RNA (mRNA) vaccines in patients with autoimmune inflammatory rheumatic diseases (AIIRD) are limited. METHODS A multicentre observational study evaluated the immunogenicity and safety of the two-dose regimen BNT162b2 mRNA vaccine in adult patients with AIIRD (n=686) compared with the general population (n=121). Serum IgG antibody levels against SARS-CoV-2 spike S1/S2 proteins were measured 2-6 weeks after the second vaccine dose. Seropositivity was defined as IgG ≥15 binding antibody units (BAU)/mL. Vaccination efficacy, safety, and disease activity were assessed within 6 weeks after the second vaccine dose. RESULTS Following vaccination, the seropositivity rate and S1/S2 IgG levels were significantly lower among patients with AIIRD versus controls (86% (n=590) vs 100%, p<0.0001 and 132.9±91.7 vs 218.6±82.06 BAU/mL, p<0.0001, respectively). Risk factors for reduced immunogenicity included older age and treatment with glucocorticoids, rituximab, mycophenolate mofetil (MMF), and abatacept. Rituximab was the main cause of a seronegative response (39% seropositivity). There were no postvaccination symptomatic cases of COVID-19 among patients with AIIRD and one mild case in the control group. Major adverse events in patients with AIIRD included death (n=2) several weeks after the second vaccine dose, non-disseminated herpes zoster (n=6), uveitis (n=2), and pericarditis (n=1). Postvaccination disease activity remained stable in the majority of patients. CONCLUSION mRNA BNTb262 vaccine was immunogenic in the majority of patients with AIIRD, with an acceptable safety profile. Treatment with glucocorticoids, rituximab, MMF, and abatacept was associated with a significantly reduced BNT162b2-induced immunogenicity.
Collapse
Affiliation(s)
- Victoria Furer
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Tali Eviatar
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Devy Zisman
- Rheumatology Unit, Carmel Hospital, Haifa, Israel
- Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Hagit Peleg
- Rheumatology Unit, Hadassah University Hospital, Jerusalem, Israel
| | - Daphna Paran
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - David Levartovsky
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Michael Zisapel
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ofir Elalouf
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Ilana Kaufman
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Roni Meidan
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
- Internal Medicine, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Adi Broyde
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Ari Polachek
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Jonathan Wollman
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Ira Litinsky
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Katya Meridor
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Hila Nochomovitz
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Adi Silberman
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Dana Rosenberg
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Joy Feld
- Rheumatology Unit, Carmel Hospital, Haifa, Israel
| | - Amir Haddad
- Rheumatology Unit, Carmel Hospital, Haifa, Israel
| | - Tal Gazzit
- Rheumatology Unit, Carmel Hospital, Haifa, Israel
| | - Muna Elias
- Rheumatology Unit, Carmel Hospital, Haifa, Israel
| | - Nizar Higazi
- Rheumatology Unit, Carmel Hospital, Haifa, Israel
| | - Fadi Kharouf
- Rheumatology Unit, Hadassah University Hospital, Jerusalem, Israel
- The Hebrew University of Jerusalem Faculty of Medicine, Jerusalem, Jerusalem, Israel
| | - Gabi Shefer
- Endocrinology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Orly Sharon
- Endocrinology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Sara Pel
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Sharon Nevo
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ori Elkayam
- Rheumatology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| |
Collapse
|
42
|
Diks AM, Overduin LA, van Leenen LD, Slobbe L, Jolink H, Visser LG, van Dongen JJM, Berkowska MA. B-Cell Immunophenotyping to Predict Vaccination Outcome in the Immunocompromised - A Systematic Review. Front Immunol 2021; 12:690328. [PMID: 34557188 PMCID: PMC8452967 DOI: 10.3389/fimmu.2021.690328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/13/2021] [Indexed: 11/13/2022] Open
Abstract
Vaccination is the most effective measure to prevent infections in the general population. Its efficiency strongly depends on the function and composition of the immune system. If the immune system lacks critical components, patients will not be fully protected despite a completed vaccination schedule. Antigen-specific serum immunoglobulin levels are broadly used correlates of protection. These are the products of terminally differentiated B cells - plasma cells. Here we reviewed the literature on how aberrancies in B-cell composition and function influence immune responses to vaccinations. In a search through five major literature databases, 6,537 unique articles published from 2000 and onwards were identified. 75 articles were included along three major research lines: extremities of life, immunodeficiency and immunosuppression. Details of the protocol can be found in the International Prospective Register of Systematic Reviews [PROSPERO (registration number CRD42021226683)]. The majority of articles investigated immune responses in adults, in which vaccinations against pneumococci and influenza were strongly represented. Lack of baseline information was the most common reason of exclusion. Irrespective of study group, three parameters measured at baseline seemed to have a predictive value in assessing vaccine efficacy: (1) distribution of B-cell subsets (mostly a reduction in memory B cells), (2) presence of exhausted/activated B cells, or B cells with an aberrant phenotype, and (3) pre-existing immunological memory. In this review we showed how pre-immunization (baseline) knowledge of circulating B cells can be used to predict vaccination efficacy. We hope that this overview will contribute to optimizing vaccination strategies, especially in immunocompromised patients.
Collapse
Affiliation(s)
- Annieck M Diks
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Lisanne A Overduin
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Department of Infectious Diseases, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Laurens D van Leenen
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Lennert Slobbe
- Department of Internal Medicine, Section of Infectious Diseases, Institute for Tropical Diseases, Erasmus Medical Center (MC), Rotterdam, Netherlands
| | - Hetty Jolink
- Department of Infectious Diseases, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Leonardus G Visser
- Department of Infectious Diseases, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | | | - Magdalena A Berkowska
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| |
Collapse
|
43
|
Perry C, Luttwak E, Balaban R, Shefer G, Morales MM, Aharon A, Tabib Y, Cohen YC, Benyamini N, Beyar-Katz O, Neaman M, Vitkon R, Keren-Khadmy N, Levin M, Herishanu Y, Avivi I. Efficacy of the BNT162b2 mRNA COVID-19 vaccine in patients with B-cell non-Hodgkin lymphoma. Blood Adv 2021; 5:3053-3061. [PMID: 34387648 PMCID: PMC8362658 DOI: 10.1182/bloodadvances.2021005094] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/04/2021] [Indexed: 01/15/2023] Open
Abstract
Patients diagnosed with B-cell non-Hodgkin lymphoma (B-NHL), particularly if recently treated with anti-CD20 antibodies, are at risk of severe COVID-19 disease. Because studies evaluating humoral response to COVID-19 vaccine in these patients are lacking, recommendations regarding vaccination strategy remain unclear. The humoral immune response to BNT162b2 messenger RNA (mRNA) COVID-19 vaccine was evaluated in patients with B-NHL who received 2 vaccine doses 21 days apart and compared with the response in healthy controls. Antibody titer, measured by the Elecsys Anti-SARS-CoV-2S assay, was evaluated 2 to 3 weeks after the second vaccine dose. Patients with B-NHL (n = 149), aggressive B-NHL (a-B-NHL; 47%), or indolent B-NHL (i-B-NHL; 53%) were evaluated. Twenty-eight (19%) were treatment naïve, 37% were actively treated with a rituximab/obinutuzumab (R/Obi)-based induction regimen or R/Obi maintenance, and 44% had last been treated with R/Obi >6 months before vaccination. A seropositive response was achieved in 89%, 7.3%, and 66.7%, respectively, with response rates of 49% in patients with B-NHL vs 98.5% in 65 healthy controls (P < .001). Multivariate analysis revealed that longer time since exposure to R/Obi and absolute lymphocyte count ≥0.9 × 103/μL predicted a positive serological response. Median time to achieve positive serology among anti-CD20 antibody-treated patients was longer in i-B-NHL vs a-B-NHL. The humoral response to BNT162b2 mRNA COVID-19 vaccine is impaired in patients with B-NHL who are undergoing R/Obi treatment. Longer time since exposure to R/Obi is associated with improved response rates to the COVID-19 vaccine. This study is registered at www.clinicaltrials.gov as #NCT04746092.
Collapse
Affiliation(s)
- C Perry
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; and
| | - E Luttwak
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; and
| | - R Balaban
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - G Shefer
- Endocrinology Laboratory, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - M M Morales
- Endocrinology Laboratory, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - A Aharon
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; and
| | - Y Tabib
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Y C Cohen
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; and
| | - N Benyamini
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; and
| | - O Beyar-Katz
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; and
| | - M Neaman
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; and
| | - R Vitkon
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; and
| | - N Keren-Khadmy
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - M Levin
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Y Herishanu
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; and
| | - I Avivi
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; and
| |
Collapse
|
44
|
Luitel P, Vais D, Gidron A. Successful Treatment of Persistent Coronavirus Disease 2019 Infection in a Patient With Hypogammaglobulinemia With REGN-COV2: A Case Report. Open Forum Infect Dis 2021; 8:ofab335. [PMID: 34405092 PMCID: PMC8344477 DOI: 10.1093/ofid/ofab335] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022] Open
Abstract
A 55-year-old man with hypogammaglobulinemia due to previous rituximab treatment developed persistent coronavirus disease 2019 pneumonia. Treatment with REGN-COV2 (casirivimab and imdevimab) resulted in the clearance of the infection. Targeted antiviral antibodies may be an important weapon in the management of immunocompromised patients infected with severe acute respiratory syndrome coronavirus 2 who fail to mount an immune response.
Collapse
Affiliation(s)
- Pankaj Luitel
- Department of Internal Medicine, Amita Saint Francis Hospital Evanston, Evanston, Illinois, USA
| | - Dana Vais
- Department of Infectious Disease, Amita Saint Mary and Elizabeth Hospital Chicago, Chicago, Illinois, USA
| | - Adi Gidron
- Department of Hematology-Oncology, Amita Saint Francis Hospital Evanston, Evanston, Illinois, USA
| |
Collapse
|
45
|
Abstract
Worldwide about one million patients are given anti-CD20 antibodies such as rituximab (RTX) for the treatment of B cell-associated diseases. Despite the success of this first therapeutic antibody, little is known about the function of its target. The role of CD20 only becomes clear in the context of the nanoscale compartmentalization of the B lymphocyte membrane. We found that CD20 is an organizer of the IgD-class nanocluster on the B cell membrane. The loss of CD20 on human B cells results in a dissolution of the IgD-class nanocluster and a transient B cell activation inducing a B cell-to-PC differentiation. Thus, CD20 is an essential gatekeeper of a membrane nanodomain and the resting state of naive B cells. CD20 is a B cell-specific membrane protein and represents an attractive target for therapeutic antibodies. Despite widespread usage of anti-CD20 antibodies for B cell depletion therapies, the biological function of their target remains unclear. Here, we demonstrate that CD20 controls the nanoscale organization of receptors on the surface of resting B lymphocytes. CRISPR/Cas9-mediated ablation of CD20 in resting B cells resulted in relocalization and interaction of the IgM-class B cell antigen receptor with the coreceptor CD19. This receptor rearrangement led to a transient activation of B cells, accompanied by the internalization of many B cell surface marker proteins. Reexpression of CD20 restored the expression of the B cell surface proteins and the resting state of Ramos B cells. Similarly, treatment of Ramos or naive human B cells with the anti-CD20 antibody rituximab induced nanoscale receptor rearrangements and transient B cell activation in vitro and in vivo. A departure from the resting B cell state followed by the loss of B cell identity of CD20-deficient Ramos B cells was accompanied by a PAX5 to BLIMP-1 transcriptional switch, metabolic reprogramming toward oxidative phosphorylation, and a shift toward plasma cell development. Thus, anti-CD20 engagement or the loss of CD20 disrupts membrane organization, profoundly altering the fate of human B cells.
Collapse
|
46
|
Galleguillos L, Alonso R. Key points to keep in mind related to COVID-19 vaccines in people with multiple sclerosis. Mult Scler Relat Disord 2021; 54:103142. [PMID: 34298479 PMCID: PMC8284074 DOI: 10.1016/j.msard.2021.103142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 06/15/2021] [Accepted: 07/05/2021] [Indexed: 11/28/2022]
Abstract
Vaccinations are often the most effective tool against certain diseases known to mankind, and their interaction with multiple sclerosis (MS) has been discussed for decades. With rapidly accumulating numbers of cases and deaths due to COVID-19, there is a global effort to respond to this pandemic in terms of scale and speed. Different platforms are currently being used around the world for the development of best COVID-19 vaccine. While some COVID-19 vaccines have already been approved by different regulatory agencies, there is scarce data in large cohorts regarding the efficacy and security of COVID-19 vaccines in people with MS. In this short review we aimed the most important information to keep in mind regarding this topic.
Collapse
Affiliation(s)
- Lorna Galleguillos
- Neurology and Psychiatry Department, Clínica alemana, Neurology and Neurosurgery Department, Clínica Dávila, Santiago, Chile.
| | - Ricardo Alonso
- Neurology Department, Hospital Ramos Mejías, Buenos Aires, Argentina.
| |
Collapse
|
47
|
Vaccine response following anti-CD20 therapy: a systematic review and meta-analysis of 905 patients. Blood Adv 2021; 5:2624-2643. [PMID: 34152403 PMCID: PMC8216656 DOI: 10.1182/bloodadvances.2021004629] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/23/2021] [Indexed: 11/20/2022] Open
Abstract
The objective of this study was to perform a systematic review of the literature on vaccine responsiveness in patients who have received anti-CD20 therapy. PubMed and EMBASE were searched up to 4 January 2021 to identify studies of vaccine immunogenicity in patients treated with anti-CD20 therapy, including patients with hematologic malignancy or autoimmune disease. The primary outcomes were seroprotection (SP), seroconversion (SC), and/or seroresponse rates for each type of vaccine reported. As the pandemic influenza vaccine (2009 H1N1) has standardized definitions for SP and SC, and represented a novel primary antigen similar to the COVID-19 vaccine, meta-analysis was conducted for SC of studies of this vaccine. Pooled estimates, relative benefit ratios (RBs), and 95% confidence intervals (CIs) were calculated using a random-effects model. Thirty-eight studies (905 patients treated with anti-CD20 therapy) were included (19 studies of patients with hematologic malignancies). Patients on active (<3 months since last dose) anti-CD20 therapy had poor responses to all types of vaccines. The pooled estimate for SC after 1 pandemic influenza vaccine dose in these patients was 3% (95% CI, 0% to 9%), with an RB of 0.05 (95% CI, 0-0.73) compared with healthy controls and 0.22 (95% CI, 0.09-0.56) compared with disease controls. SC compared with controls seems abrogated for at least 6 months following treatment (3-6 months post anti-CD20 therapy with an RB of 0.50 [95% CI, 0.24-1.06] compared with healthy and of 0.44 [95% CI, 0.23-0.84] compared with disease controls). For all vaccine types, response to vaccination improves incrementally over time, but may not reach the level of healthy controls even 12 months after therapy.
Collapse
|
48
|
Messmer M, Wagner-Johnston N. COVID-19 vaccination in patients on rituximab: a survey of lymphoma physicians at NCI designated cancer centers. Leuk Lymphoma 2021; 62:3019-3022. [PMID: 34151698 DOI: 10.1080/10428194.2021.1941939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Marcus Messmer
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Nina Wagner-Johnston
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
49
|
Woopen C, Schleußner K, Akgün K, Ziemssen T. Approach to SARS-CoV-2 Vaccination in Patients With Multiple Sclerosis. Front Immunol 2021; 12:701752. [PMID: 34234787 PMCID: PMC8256163 DOI: 10.3389/fimmu.2021.701752] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/07/2021] [Indexed: 12/18/2022] Open
Abstract
For more than a year now, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been causing the coronavirus disease (COVID-19) pandemic with high mortality and detrimental effects on society, economy, and individual lives. Great hopes are being placed on vaccination as one of the most potent escape strategies from the pandemic and multiple vaccines are already in clinical use. However, there is still a lot of insecurity about the safety and efficacy of vaccines in patients with autoimmune diseases like multiple sclerosis (MS), especially under treatment with immunomodulatory or immunosuppressive drugs. We propose strategic approaches to SARS-CoV-2 vaccination management in MS patients and encourage fellow physicians to measure the immune response in their patients. Notably, both humoral and cellular responses should be considered since the immunological equivalent for protection from SARS-CoV-2 after infection or vaccination still remains undefined and will most likely involve antiviral cellular immunity. It is important to gain insights into the vaccine response of immunocompromised patients in order to be able to deduce sensible strategies for vaccination in the future.
Collapse
Affiliation(s)
| | | | | | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus Dresden, Technical University of Dresden, Dresden, Germany
| |
Collapse
|
50
|
Mason A, Anver H, Lwin M, Holroyd C, Faust SN, Edwards CJ. Lupus, vaccinations and COVID-19: What we know now. Lupus 2021; 30:1541-1552. [PMID: 34134555 DOI: 10.1177/09612033211024355] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the virus causing Coronavirus disease 2019 (COVID-19), has had a huge impact on health services, with a high mortality associated with complications including pneumonia and acute respiratory distress syndrome. Patients with systemic lupus erythematosus (SLE) are at increased risk of viral infections, and recent data suggests they may be at an increased risk of poor outcomes with COVID-19. This may be particularly true for those on rituximab or high dose steroids. A huge international effort from the scientific community has so far resulted in the temporary authorisation of three vaccines which offer protection against SARS-CoV-2, with over 30 other vaccines being evaluated in ongoing trials. Although there has historically been concern that vaccines may trigger disease flares of SLE, there is little convincing evidence to show this. In general lupus patients appear to gain good protection from vaccination, although there may be reduced efficacy in those with high disease activity or those on immunosuppressive therapies, such as rituximab or high dose steroids. Recent concerns have been raised regarding rare clotting events with the AstraZeneca/Oxford vaccine and it is currently unknown whether this risk is higher for those patients with secondary antiphospholipid syndrome. With the possibility of annual COVID vaccination programmes in the future, prospective data collection and registries looking at the effect of vaccination on SLE disease control, the incidence of COVID-19 in SLE patients and severity of COVID-19 disease course would all be useful. As mass vaccination programmes begin to roll out across the world, we assess the evidence of the use of vaccines in SLE patients and in particular vaccines targeting SARS-CoV-2.
Collapse
Affiliation(s)
- Alice Mason
- Rheumatology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Himashi Anver
- Rheumatology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - May Lwin
- NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Christopher Holroyd
- Rheumatology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Saul N Faust
- NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Christopher J Edwards
- Rheumatology, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| |
Collapse
|