1
|
Bazile C, Abdel Malik MM, Ackeifi C, Anderson RL, Beck RW, Donath MY, Dutta S, Hedrick JA, Karpen SR, Kay TWH, Marder T, Marinac M, McVean J, Meyer R, Pettus J, Quattrin T, Verstegen RHJ, Vieth JA, Latres E. TNF-α inhibitors for type 1 diabetes: exploring the path to a pivotal clinical trial. Front Immunol 2024; 15:1470677. [PMID: 39411715 PMCID: PMC11473295 DOI: 10.3389/fimmu.2024.1470677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/11/2024] [Indexed: 10/19/2024] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by the destruction of insulin-producing β-cells in the pancreas. This destruction leads to chronic hyperglycemia, necessitating lifelong insulin therapy to manage blood glucose levels. Typically diagnosed in children and young adults, T1D can, however, occur at any age. Ongoing research aims to uncover the precise mechanisms underlying T1D and to develop potential interventions. These include efforts to modulate the immune system, regenerate β-cells, and create advanced insulin delivery systems. Emerging therapies, such as closed-loop insulin pumps, stem cell-derived β-cell replacement and disease-modifying therapies (DMTs), offer hope for improving the quality of life for individuals with T1D and potentially moving towards a cure. Currently, there are no disease-modifying therapies approved for stage 3 T1D. Preserving β-cell function in stage 3 T1D is associated with better clinical outcomes, including lower HbA1c and decreased risk of hypoglycemia, neuropathy, and retinopathy. Tumor Necrosis Factor alpha (TNF-α) inhibitors have demonstrated efficacy at preserving β-cell function by measurement of C-peptide in two clinical trials in people with stage 3 T1D. However, TNF-α inhibitors have yet to be evaluated in a pivotal trial for T1D. To address the promising clinical findings of TNF-α inhibitors in T1D, Breakthrough T1D convened a panel of key opinion leaders (KOLs) in the field. The workshop aimed to outline an optimal clinical path for moving TNF-α inhibitors to a pivotal clinical trial in T1D. Here, we summarize the evidence for the beneficial use of TNF-α inhibitors in T1D and considerations for strategies collectively identified to advance TNF-α inhibitors beyond phase 2 clinical studies for stage 3 T1D.
Collapse
Affiliation(s)
- Cassandra Bazile
- Breakthrough T1D (formerly known as JDRF), New York,
NY, United States
| | | | - Courtney Ackeifi
- Breakthrough T1D (formerly known as JDRF), New York,
NY, United States
| | | | - Roy W. Beck
- Jaeb Center for Health Research, Tampa, FL,
United States
| | - Marc Y. Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University of Basel, Basel, Switzerland
| | - Sanjoy Dutta
- Breakthrough T1D (formerly known as JDRF), New York,
NY, United States
| | | | - Stephen R. Karpen
- Breakthrough T1D (formerly known as JDRF), New York,
NY, United States
| | - Thomas W. H. Kay
- St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
| | | | - Marjana Marinac
- Breakthrough T1D (formerly known as JDRF), New York,
NY, United States
| | | | | | - Jeremy Pettus
- Division of Endocrinology and Metabolism, Department of Medicine, University of
California San Diego, La Jolla, CA, United States
| | - Teresa Quattrin
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Ruud H. J. Verstegen
- Division of Clinical Pharmacology and Toxicology, Department of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
- Division of Rheumatology, Department of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
| | - Joshua A. Vieth
- Breakthrough T1D (formerly known as JDRF), New York,
NY, United States
| | - Esther Latres
- Breakthrough T1D (formerly known as JDRF), New York,
NY, United States
| |
Collapse
|
2
|
Nijhuis L, Swart JF, Prakken BJ, van Loosdregt J, Vastert SJ. The clinical and experimental treatment of Juvenile Idiopathic Arthritis. Clin Exp Immunol 2023; 213:276-287. [PMID: 37074076 PMCID: PMC10571000 DOI: 10.1093/cei/uxad045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/02/2023] [Accepted: 04/18/2023] [Indexed: 04/20/2023] Open
Abstract
Juvenile idiopathic arthritis (JIA) is the most common chronic rheumatic disease in children and comprises of multiple subtypes. The most relevant disease subtypes, grouped upon current insight in disease mechanisms, are nonsystemic (oligo- and polyarticular) JIA and systemic JIA (sJIA). In this review, we summarize some of the main proposed mechanisms of disease in both nonsystemic and sJIA and discuss how current therapeutic modalities target some of the pathogenic immune pathways. Chronic inflammation in nonsystemic JIA is the result of a complex interplay between effector and regulatory immune cell subsets, with adaptive immune cells, specifically T-cell subsets and antigen-presenting cells, in a central role. There is, however, also innate immune cell contribution. SJIA is nowadays recognized as an acquired chronic inflammatory disorder with striking autoinflammatory features in the first phase of the disease. Some sJIA patients develop a refractory disease course, with indications for involvement of adaptive immune pathways as well. Currently, therapeutic strategies are directed at suppressing effector mechanisms in both non-systemic and sJIA. These strategies are often not yet optimally tuned nor timed to the known active mechanisms of disease in individual patients in both non-systemic and sJIA. We discuss current treatment strategies in JIA, specifically the 'Step-up' and 'Treat to Target approach' and explore how increased insight into the biology of disease may translate into future more targeted strategies for this chronic inflammatory disease at relevant time points: preclinical disease, active disease, and clinically inactive disease.
Collapse
Affiliation(s)
- L Nijhuis
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of pediatric rheumatology & immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J F Swart
- Department of pediatric rheumatology & immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- University of Utrecht, Utrecht, The Netherlands
| | - B J Prakken
- Department of pediatric rheumatology & immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- University of Utrecht, Utrecht, The Netherlands
| | - J van Loosdregt
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- University of Utrecht, Utrecht, The Netherlands
| | - S J Vastert
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of pediatric rheumatology & immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- University of Utrecht, Utrecht, The Netherlands
| |
Collapse
|
3
|
Long AM, Marston B. Juvenile Idiopathic Arthritis. Pediatr Rev 2023; 44:565-577. [PMID: 37777651 DOI: 10.1542/pir.2022-005623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/02/2023]
Abstract
Juvenile idiopathic arthritis (JIA) comprises a group of heterogenous disorders characterized by childhood-onset chronic joint inflammation. It is the most common rheumatologic disease in the pediatric population and an important cause of chronic illness in children. Early recognition and treatment are vital to prevent sequelae of uncontrolled inflammation on the developing skeleton. JIA can have significant complications that general pediatricians should be aware of, especially uveitis, which can be insidious and asymptomatic in very young children, and macrophage activation syndrome, which can be life-threatening if not recognized and appropriately treated. Although advances have been made in the past few decades, the etiology of JIA remains incompletely understood. Efforts are underway to refine the classification of JIA. The currently accepted classification scheme identifies subsets of JIA that are important clinically in terms of prognosis and tailoring treatment approaches. However, it is limited in identifying homogenous groups of children with early childhood onset and antinuclear antibody positivity, which may have different pathogenic mechanisms that could be important in developing more targeted and effective treatment approaches in the future. Treatment strategies for JIA have changed significantly in recent years with the availability of multiple newer targeted therapies, often modeled after medications used in adult-onset forms of arthritis. These treatments, and likely many others to come, have markedly improved symptom control and reduced complications in patients with JIA.
Collapse
Affiliation(s)
- Andrew M Long
- Division of Pediatric Rheumatology, Golisano Children's Hospital, Rochester, NY
- Division of Allergy, Immunology, and Rheumatology, University of Rochester Medical Center, Rochester, NY
| | - Bethany Marston
- Division of Pediatric Rheumatology, Golisano Children's Hospital, Rochester, NY
- Division of Allergy, Immunology, and Rheumatology, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
4
|
Tomé C, Oliveira-Ramos F, Campanilho-Marques R, Mourão AF, Sousa S, Marques C, Melo AT, Teixeira RL, Martins AP, Moeda S, Costa-Reis P, Torres RP, Bandeira M, Fonseca H, Gonçalves M, Santos MJ, Graca L, Fonseca JE, Moura RA. Children with extended oligoarticular and polyarticular juvenile idiopathic arthritis have alterations in B and T follicular cell subsets in peripheral blood and a cytokine profile sustaining B cell activation. RMD Open 2023; 9:e002901. [PMID: 37652558 PMCID: PMC10476142 DOI: 10.1136/rmdopen-2022-002901] [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: 11/28/2022] [Accepted: 07/03/2023] [Indexed: 09/02/2023] Open
Abstract
OBJECTIVES The main goal of this study was to characterise the frequency and phenotype of B, T follicular helper (Tfh) and T follicular regulatory (Tfr) cells in peripheral blood and the cytokine environment present in circulation in children with extended oligoarticular juvenile idiopathic arthritis (extended oligo JIA) and polyarticular JIA (poly JIA) when compared with healthy controls, children with persistent oligoarticular JIA (persistent oligo JIA) and adult JIA patients. METHODS Blood samples were collected from 105 JIA patients (children and adults) and 50 age-matched healthy individuals. The frequency and phenotype of B, Tfh and Tfr cells were evaluated by flow cytometry. Serum levels of APRIL, BAFF, IL-1β, IL-2, IL-4, IL-6, IL-10, IL-17A, IL-21, IL-22, IFN-γ, PD-1, PD-L1, sCD40L, CXCL13 and TNF were measured by multiplex bead-based immunoassay and/or ELISA in all groups included. RESULTS The frequency of B, Tfh and Tfr cells was similar between JIA patients and controls. Children with extended oligo JIA and poly JIA, but not persistent oligo JIA, had significantly lower frequencies of plasmablasts, regulatory T cells and higher levels of Th17-like Tfh cells in circulation when compared with controls. Furthermore, APRIL, BAFF, IL-6 and IL-17A serum levels were significantly higher in paediatric extended oligo JIA and poly JIA patients when compared with controls. These immunological alterations were not found in adult JIA patients in comparison to controls. CONCLUSIONS Our results suggest a potential role and/or activation profile of B and Th17-like Tfh cells in the pathogenesis of extended oligo JIA and poly JIA, but not persistent oligo JIA.
Collapse
Affiliation(s)
- Catarina Tomé
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Filipa Oliveira-Ramos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, EPE, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Raquel Campanilho-Marques
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, EPE, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Ana F Mourão
- Rheumatology Department, Hospital de São Francisco Xavier, Centro Hospitalar Lisboa Ocidental, EPE, Lisbon, Portugal
| | - Sandra Sousa
- Reumatology Department, Hospital Garcia de Orta, EPE, Almada, Portugal
| | - Cláudia Marques
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Ana T Melo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, EPE, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Rui L Teixeira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, EPE, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Ana P Martins
- Pediatric Surgery Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, EPE, Lisbon, Portugal
| | - Sofia Moeda
- Department of Pediatrics, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, EPE, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Patrícia Costa-Reis
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Department of Pediatrics, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, EPE, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Rita P Torres
- Rheumatology Department, Hospital de São Francisco Xavier, Centro Hospitalar Lisboa Ocidental, EPE, Lisbon, Portugal
| | - Matilde Bandeira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, EPE, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Helena Fonseca
- Department of Pediatrics, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, EPE, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Miroslava Gonçalves
- Pediatric Surgery Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, EPE, Lisbon, Portugal
| | - Maria J Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Reumatology Department, Hospital Garcia de Orta, EPE, Almada, Portugal
| | - Luis Graca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - João E Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, EPE, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Rita A Moura
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| |
Collapse
|
5
|
Athni TS, Barmettler S. Hypogammaglobulinemia, late-onset neutropenia, and infections following rituximab. Ann Allergy Asthma Immunol 2023; 130:699-712. [PMID: 36706910 PMCID: PMC10247428 DOI: 10.1016/j.anai.2023.01.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/23/2022] [Accepted: 01/16/2023] [Indexed: 01/26/2023]
Abstract
Rituximab is a chimeric anti-CD20 monoclonal antibody that targets CD20-expressing B lymphocytes, has a well-defined efficacy and safety profile, and is broadly used to treat a wide array of diseases. In this review, we cover the mechanism of action of rituximab and focus on hypogammaglobulinemia and late-onset neutropenia-2 immune effects secondary to rituximab-and subsequent infection. We review risk factors and highlight key considerations for immunologic monitoring and clinical management of rituximab-induced secondary immune deficiencies. In patients treated with rituximab, monitoring for hypogammaglobulinemia and infections may help to identify the subset of patients at high risk for developing poor B cell reconstitution, subsequent infections, and adverse complications. These patients may benefit from early interventions such as vaccination, antibacterial prophylaxis, and immunoglobulin replacement therapy. Systematic evaluation of immunoglobulin levels and peripheral B cell counts by flow cytometry, both at baseline and periodically after therapy, is recommended for monitoring. In addition, in those patients with prolonged hypogammaglobulinemia and increased infections after rituximab use, immunologic evaluation for inborn errors of immunity may be warranted to further risk stratification, increase monitoring, and assist in therapeutic decision-making. As the immunologic effects of rituximab are further elucidated, personalized approaches to minimize the risk of adverse reactions while maximizing benefit will allow for improved care of patients with decreased morbidity and mortality.
Collapse
Affiliation(s)
| | - Sara Barmettler
- Allergy and Clinical Immunology Unit, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
6
|
Julé AM, Lam KP, Taylor M, Hoyt KJ, Wei K, Gutierrez-Arcelus M, Case SM, Chandler M, Chang MH, Cohen EM, Dedeoglu F, Halyabar O, Hausmann J, Hazen MM, Janssen E, Lo J, Lo MS, Meidan E, Roberts JE, Wobma H, Son MBF, Sundel RP, Lee PY, Sage PT, Chatila TA, Nigrovic PA, Rao DA, Henderson LA. Disordered T cell-B cell interactions in autoantibody-positive inflammatory arthritis. Front Immunol 2023; 13:1068399. [PMID: 36685593 PMCID: PMC9849554 DOI: 10.3389/fimmu.2022.1068399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023] Open
Abstract
T peripheral helper (Tph) cells, identified in the synovium of adults with seropositive rheumatoid arthritis, drive B cell maturation and antibody production in non-lymphoid tissues. We sought to determine if similarly dysregulated T cell-B cell interactions underlie another form of inflammatory arthritis, juvenile oligoarthritis (oligo JIA). Clonally expanded Tph cells able to promote B cell antibody production preferentially accumulated in the synovial fluid (SF) of oligo JIA patients with antinuclear antibodies (ANA) compared to autoantibody-negative patients. Single-cell transcriptomics enabled further definition of the Tph gene signature in inflamed tissues and showed that Tph cells from ANA-positive patients upregulated genes associated with B cell help to a greater extent than patients without autoantibodies. T cells that co-expressed regulatory T and B cell-help factors were identified. The phenotype of these Tph-like Treg cells suggests an ability to restrain T cell-B cell interactions in tissues. Our findings support the central role of disordered T cell-help to B cells in autoantibody-positive arthritides.
Collapse
Affiliation(s)
- Amélie M. Julé
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Ki Pui Lam
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Maria Taylor
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Kacie J. Hoyt
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Maria Gutierrez-Arcelus
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Siobhan M. Case
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Mia Chandler
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Margaret H. Chang
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Ezra M. Cohen
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Boston Medical Center, Boston University School of Medicine, Boston, MA, United States
| | - Fatma Dedeoglu
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Olha Halyabar
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jonathan Hausmann
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Melissa M. Hazen
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Erin Janssen
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jeffrey Lo
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Mindy S. Lo
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Esra Meidan
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jordan E. Roberts
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Holly Wobma
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Mary Beth F. Son
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Robert P. Sundel
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Pui Y. Lee
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Talal A. Chatila
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Peter A. Nigrovic
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Deepak A. Rao
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Lauren A. Henderson
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
7
|
Blincoe A, Labrosse R, Abraham RS. Acquired B-cell deficiency secondary to B-cell-depleting therapies. J Immunol Methods 2022; 511:113385. [PMID: 36372267 DOI: 10.1016/j.jim.2022.113385] [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: 06/19/2022] [Revised: 09/26/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022]
Abstract
The advantage of the newer biological therapies is that the immunosuppressive effect is targeted, in contrast, to the standard, traditional immunomodulatory agents, which have a more global effect. However, there are unintended targets and consequences, even to these "precise" therapeutics, leading to acquired or secondary immunodeficiencies. Besides depleting specific cellular immune subsets, these biological agents, which include monoclonal antibodies against biologically relevant molecules, often have broader functional immune consequences, which become apparent over time. This review focuses on acquired B-cell immunodeficiency, secondary to the use of B-cell depleting therapeutic agents. Among the many adverse consequences of B-cell depletion is the risk of hypogammaglobulinemia, failure of B-cell recovery, impaired B-cell differentiation, and risk of infections. Factors, which modulate the outcomes of B-cell depleting therapies, include the intrinsic nature of the underlying disease, the concomitant use of other immunomodulatory agents, and the clinical status of the patient and other co-existing morbidities. This article seeks to explore the mechanism of action of B-cell depleting agents, the clinical utility and adverse effects of these therapies, and the relevance of systematic and serial laboratory immune monitoring in identifying patients at risk for developing immunological complications, and who may benefit from early intervention to mitigate the secondary consequences. Though these biological drugs are gaining widespread use, a harmonized approach to immune evaluation pre-and post-treatment has not yet gained traction across multiple clinical specialties, because of which, the true prevalence of these adverse events cannot be determined in the treated population, and a systematic and evidence-based dosing schedule cannot be developed. The aim of this review is to bring these issues into focus, and initiate a multi-specialty, data-driven approach to immune monitoring.
Collapse
Affiliation(s)
- Annaliesse Blincoe
- Department of Paediatric Immunology and Allergy, Starship Child Health, Auckland, NZ, New Zealand
| | - Roxane Labrosse
- Department of Pediatrics, CHU Sainte-Justine, University of Montreal, Montreal, Canada
| | - Roshini S Abraham
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
8
|
Moura RA, Fonseca JE. B Cells on the Stage of Inflammation in Juvenile Idiopathic Arthritis: Leading or Supporting Actors in Disease Pathogenesis? Front Med (Lausanne) 2022; 9:851532. [PMID: 35449805 PMCID: PMC9017649 DOI: 10.3389/fmed.2022.851532] [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: 01/10/2022] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Juvenile idiopathic arthritis (JIA) is a term that collectively refers to a group of chronic childhood arthritides, which together constitute the most common rheumatic condition in children. The International League of Associations for Rheumatology (ILAR) criteria define seven categories of JIA: oligoarticular, polyarticular rheumatoid factor (RF) negative (RF-), polyarticular RF positive (RF+), systemic, enthesitis-related arthritis, psoriatic arthritis, and undifferentiated arthritis. The ILAR classification includes persistent and extended oligoarthritis as subcategories of oligoarticular JIA, but not as distinct categories. JIA is characterized by a chronic inflammatory process affecting the synovia that begins before the age of 16 and persists at least 6 weeks. If not treated, JIA can cause significant disability and loss of quality of life. Treatment of JIA is adjusted according to the severity of the disease as combinations of non-steroidal anti-inflammatory drugs (NSAIDs), synthetic and/ or biological disease modifying anti-rheumatic drugs (DMARDs). Although the disease etiology is unknown, disturbances in innate and adaptive immune responses have been implicated in JIA development. B cells may have important roles in JIA pathogenesis through autoantibody production, antigen presentation, cytokine release and/ or T cell activation. The study of B cells has not been extensively explored in JIA, but evidence from the literature suggests that B cells might have indeed a relevant role in JIA pathophysiology. The detection of autoantibodies such as antinuclear antibodies (ANA), RF and anti-citrullinated protein antibodies (ACPA) in JIA patients supports a breakdown in B cell tolerance. Furthermore, alterations in B cell subpopulations have been documented in peripheral blood and synovial fluid from JIA patients. In fact, altered B cell homeostasis, B cell differentiation and B cell hyperactivity have been described in JIA. Of note, B cell depletion therapy with rituximab has been shown to be an effective and well-tolerated treatment in children with JIA, which further supports B cell intervention in disease development.
Collapse
Affiliation(s)
- Rita A Moura
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João Eurico Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon Academic Medical Centre, Lisbon, Portugal
| |
Collapse
|
9
|
Abstract
Juvenile idiopathic arthritis (JIA) is an umbrella term for arthritis of unknown origin, lasting for >6 weeks with onset before 16 years of age. JIA is the most common chronic inflammatory rheumatic condition of childhood. According to the International League Against Rheumatism (ILAR) classification, seven mutually exclusive categories of JIA exist based on disease manifestations during the first 6 months of disease. Although the ILAR classification has been useful to foster research, it has been criticized mainly as it does not distinguish those forms of chronic arthritis observed in adults and in children from those that may be unique to childhood. Hence, efforts to provide a new evidence-based classification are ongoing. Similar to arthritis observed in adults, pathogenesis involves autoimmune and autoinflammatory mechanisms. The field has witnessed a remarkable improvement in therapeutic possibilities of JIA owing to the availability of new potent drugs and the possibility to perform controlled trials with support from legislative interventions and large networks availability. The goal of drug therapy in JIA is to rapidly reduce disease activity to inactive disease or clinical remission, minimize drug side effects and achieve a quality of life comparable to that of healthy peers. As JIA can influence all aspects of a child's and their family's life, researchers increasingly recognize improvement of health-related quality of life as a key treatment goal.
Collapse
|
10
|
Rosina S, Natoli V, Santaniello S, Trincianti C, Consolaro A, Ravelli A. Novel biomarkers for prediction of outcome and therapeutic response in juvenile idiopathic arthritis. Expert Rev Clin Immunol 2021; 17:853-870. [PMID: 34139935 DOI: 10.1080/1744666x.2021.1945441] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The search for biomarkers in juvenile idiopathic arthritis (JIA) is a promising and rapidly expanding field of investigation. The biomarkers identified so far may help to dissect the clinical heterogeneity of the illness, measure the level of disease activity, predict clinical remission, relapse, response to medications, course over time, complications, and forestall disease flares. AREAS COVERED We provide a summary of the most recent advances in the development and application of biomarkers in JIA. We performed a PubMed search for significant articles combining predetermined keywords related to biomarkers in non-systemic and systemic JIA, chronic uveitis, and macrophage activation syndrome (MAS). The biomarkers available or under study are presented and discussed separately for non-systemic and systemic subtypes and for the two main disease complications, uveitis and MAS. EXPERT OPINION The incorporation of valid and reliable biomarkers in standard clinical care may help to design better patient-tailored treatment regimens and to improve the therapeutic strategies based on the treat-to-target approach. The establishment of biomarkers that predict the risk of disease flare may lead to define the optimal modalities for treatment discontinuation after the achievement of clinical remission.
Collapse
Affiliation(s)
- Silvia Rosina
- UOC Clinica Pediatrica E Reumatologia, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Valentina Natoli
- Dipartimento Di Neuroscienze, Riabilitazione, Oftalmologia, Genetica E Scienze Materno-Infantili (Dinogmi), Università Degli Studi Di Genova, Genoa, Italy
| | - Stefania Santaniello
- Dipartimento Di Neuroscienze, Riabilitazione, Oftalmologia, Genetica E Scienze Materno-Infantili (Dinogmi), Università Degli Studi Di Genova, Genoa, Italy
| | - Chiara Trincianti
- Dipartimento Di Neuroscienze, Riabilitazione, Oftalmologia, Genetica E Scienze Materno-Infantili (Dinogmi), Università Degli Studi Di Genova, Genoa, Italy
| | - Alessandro Consolaro
- UOC Clinica Pediatrica E Reumatologia, IRCCS Istituto Giannina Gaslini, Genoa, Italy.,Dipartimento Di Neuroscienze, Riabilitazione, Oftalmologia, Genetica E Scienze Materno-Infantili (Dinogmi), Università Degli Studi Di Genova, Genoa, Italy
| | - Angelo Ravelli
- UOC Clinica Pediatrica E Reumatologia, IRCCS Istituto Giannina Gaslini, Genoa, Italy.,Dipartimento Di Neuroscienze, Riabilitazione, Oftalmologia, Genetica E Scienze Materno-Infantili (Dinogmi), Università Degli Studi Di Genova, Genoa, Italy.,Sechenov First Moscow State Medical University, Moscow, Russian Federation
| |
Collapse
|
11
|
Fischer J, Dirks J, Klaussner J, Haase G, Holl-Wieden A, Hofmann C, Hackenberg S, Girschick H, Morbach H. Clonally expanded PD-1 hi CXCR5 - CD4 + peripheral T helper cells promote differentiation of CD21 lo/- CD11c + double negative B cells in the joints of ANA+ JIA patients. Arthritis Rheumatol 2021; 74:150-162. [PMID: 34196496 DOI: 10.1002/art.41913] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/18/2021] [Accepted: 06/29/2021] [Indexed: 11/10/2022]
Abstract
OBJECTIVES Antinuclear antibody (ANA) positive Juvenile Idiopathic Arthritis (JIA) is characterized by synovial B cell hyperactivity, but the precise role of CD4+ T cells in promoting local B cell activation is unknown. The objective of this study is to unravel the phenotype and function of synovial CD4+ T cells that promote the aberrant B cell activation in JIA. METHODS Flow cytometric analysis was performed to compare the phenotype and cytokine pattern of synovial fluid (SF) PD-1hi CD4+ T cells with tonsil TFH cells. TCRVB next generation sequencing was applied to analyze T cell subsets for signs of clonal expansion. The functional impact of these T cell subsets on B cells was dissected in in vitro co-cultures. RESULTS Multidimensional flow-cytometric analysis revealed the expansion of IL-21 and IFN-γ co-expressing PD-1hi CXCR5- HLA-DR+ CD4+ T cells that accumulate in the joints of ANA+ JIA patients. These T cells exhibited signs of clonal expansion with restricted TCR clonotypes. Phenotypically they resembled peripheral T helper (TPH ) cells with an extrafollicular chemokine receptor pattern and high T-bet and Blimp-1 but low Bcl-6 expression. SF TPH cells particularly skewed B cell differentiation towards a CD21lo/- CD11c+ phenotype by provision of IL-21 and IFN-γ in vitro and correlated with the appearance of SF CD21lo/- CD11c+ CD27- IgM- double-negative B cells (BDN ) in situ. CONCLUSION Clonally expanded CD4+ TPH cells accumulate in the joints of ANA+ JIA patients and particularly promote CD21lo/- CD11c+ BDN cell differentiation The expansion of TPH and BDN cells might reflect the autoimmune response present in the joints of ANA+ JIA patients.
Collapse
Affiliation(s)
- Jonas Fischer
- Pediatric Immunology, Department of Pediatrics, University of Würzburg, Würzburg, Germany
| | - Johannes Dirks
- Pediatric Immunology, Department of Pediatrics, University of Würzburg, Würzburg, Germany
| | - Julia Klaussner
- Pediatric Immunology, Department of Pediatrics, University of Würzburg, Würzburg, Germany
| | - Gabriele Haase
- Pediatric Immunology, Department of Pediatrics, University of Würzburg, Würzburg, Germany
| | - Annette Holl-Wieden
- Pediatric Rheumatology and Osteology, Department of Pediatrics, University of Würzburg, Würzburg, Germany
| | - Christine Hofmann
- Pediatric Rheumatology and Osteology, Department of Pediatrics, University of Würzburg, Würzburg, Germany
| | - Stephan Hackenberg
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Würzburg, Würzburg, Germany
| | - Hermann Girschick
- Children's Hospital, Vivantes Klinikum im Friedrichshain, Berlin, Germany
| | - Henner Morbach
- Pediatric Immunology, Department of Pediatrics, University of Würzburg, Würzburg, Germany.,Pediatric Rheumatology and Osteology, Department of Pediatrics, University of Würzburg, Würzburg, Germany
| |
Collapse
|
12
|
Choida V, Hall-Craggs M, Jebson BR, Fisher C, Leandro M, Wedderburn LR, Ciurtin C. Biomarkers of Response to Biologic Therapy in Juvenile Idiopathic Arthritis. Front Pharmacol 2021; 11:635823. [PMID: 33603671 PMCID: PMC7884612 DOI: 10.3389/fphar.2020.635823] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/31/2020] [Indexed: 01/22/2023] Open
Abstract
Background: Juvenile idiopathic arthritis (JIA) is the most common chronic inflammatory arthritis of childhood, characterized by various clinical phenotypes associated with variable prognosis. Significant progress has been achieved with the use of biologic treatments, which specifically block pro-inflammatory molecules involved in the disease pathogenesis. The most commonly used biologics in JIA are monoclonal antibodies and recombinant proteins targeting interleukins 1 (IL-1) and 6 (IL-6), and tumor necrosis factor α (TNF-α). Several biomarkers have been investigated in JIA. Aims: To assess the level of evidence available regarding the role of biomarkers in JIA related to guiding clinical and therapeutic decisions, providing disease prognostic information, facilitating disease activity monitoring and assessing biologic treatment response in JIA, as well as propose new strategies for biologic therapy-related biomarker use in JIA. Methods: We searched PubMed for relevant literature using predefined key words corresponding to several categories of biomarkers to assess their role in predicting and assessing biologic treatment response and clinical remission in JIA. Results: We reviewed serological, cellular, genetic, transcriptomic and imaging biomarkers, to identify candidates that are both well-established and widely used, as well as newly investigated in JIA on biologic therapy. We evaluated their role in management of JIA as well as identified the unmet needs for new biomarker discovery and better clinical applications. Conclusion: Although there are no ideal biomarkers in JIA, we identified serological biomarkers with potential clinical utility. We propose strategies of combining biomarkers of response to biologics in JIA, as well as routine implementation of clinically acceptable imaging biomarkers for improved disease assessment performance.
Collapse
Affiliation(s)
- Varvara Choida
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, Division of Medicine, University College London, London, United Kingdom
- Department of Adolescent Rheumatology, University College London Hospital, London, United Kingdom
| | | | - Bethany R. Jebson
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, Division of Medicine, University College London, London, United Kingdom
- University College London Great Ormond Street Institute for Child Health, London, United Kingdom
| | - Corinne Fisher
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, Division of Medicine, University College London, London, United Kingdom
- Department of Adolescent Rheumatology, University College London Hospital, London, United Kingdom
| | - Maria Leandro
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, Division of Medicine, University College London, London, United Kingdom
- Department of Adolescent Rheumatology, University College London Hospital, London, United Kingdom
| | - Lucy R. Wedderburn
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, Division of Medicine, University College London, London, United Kingdom
- University College London Great Ormond Street Institute for Child Health, London, United Kingdom
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, United Kingdom
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, Division of Medicine, University College London, London, United Kingdom
- Department of Adolescent Rheumatology, University College London Hospital, London, United Kingdom
| |
Collapse
|
13
|
Nicácio AAMF, Peracchi OAB, Yamada J, Fraga MM, Vitalle MS, de Moraes-Pinto MI, Terreri MT. Tdap booster to adolescents with juvenile idiopathic arthritis on and off anti-TNF agents is safe and immunogenic. Vaccine 2021; 39:1165-1172. [PMID: 33478788 DOI: 10.1016/j.vaccine.2020.12.071] [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: 07/05/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 10/22/2022]
Abstract
INTRODUCTION The response to vaccines in juvenile idiopathic arthritis (JIA) patients on and off anti-tumor necrosis factor (anti-TNF) agents remains highly discussed. There are no published studies on the immune response following a Tdap booster dose in JIA patients so far. OBJECTIVE To evaluate the immune response and safety after a Tdap booster in JIA patients and in healthy adolescents. METHODS Nineteen adolescents with JIA according to the ILAR criteria on anti-TNF medication, 19 adolescents with JIA off anti-TNF medication, and 27 healthy adolescents (control group) were compared after a Tdap booster. Adverse events and disease activity were evaluated. Lymphocyte immunophenotyping was performed by flow cytometry. Tetanus, diphtheria and pertussis toxin antibodies were assessed by ELISA; whole blood was stimulated with whole-cell pertussis, and supernatants were assessed for cytokines by xMAP. RESULTS The three groups showed a similar frequency of adverse events. There was no disease reactivation after the Tdap booster. Tetanus, diphtheria and pertussis antibodies showed a significant response when D0 and D14 concentrations were compared in both JIA groups and controls. Over time, a different pattern of response to the Tdap booster was observed among the groups for tetanus antibodies (p = 0.005) but not for diphtheria and pertussis antibodies. In contrast to the protection attained for tetanus and diphtheria, in the three groups, not all individuals showed pertussis seroconversion at either D14 or D28. In addition, the seroconversion of three subjects with JIA on anti-TNF medication was not maintained at D28. JIA patients off anti-TNF showed a higher percentage of naive CD8 + T cells (p = 0.007) and central memory CD8 + cells (p = 0.003) and a lower percentage of effector CD8 + T cells (p = 0.003) and NK cell numbers (p = 0.018) than the control group. The JIA group off anti-TNF medication had fewer B lymphocytes than both the JIA group on anti-TNF medication and the control group (p = 0.016). Cellular immunity to Bordetella pertussis showed that IFNγ levels were significantly lower in both JIA groups than in the control group (p = 0.003), IL10 levels were higher in the JIA off anti-TNF group (p = 0.009), IL17A and IL5 levels were lower in the JIA on anti-TNF group than in the control group (p = 0.018 and p = 0.016, respectively); however, an increase in IFNγ (p = 0.008), IL17A (p = 0.030) and TNFα (p = 0.041) levels was observed at D14 in both patient groups. Both JIA groups showed higher levels of IL21 than the control group (p = 0.023). CONCLUSION We conclude that individuals with JIA on or off anti-TNF agents showed a good response to a booster dose for the three antigens studied in the absence of major adverse events and without the reactivation of the disease.
Collapse
Affiliation(s)
- Aline Alencar M F Nicácio
- Unit of Pediatric Rheumatology, Department of Pediatrics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Octávio A B Peracchi
- Unit of Pediatric Rheumatology, Department of Pediatrics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Juliana Yamada
- Research Laboratory, Division of Pediatric Infectious Diseases, Department of Pediatrics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Melissa M Fraga
- Unit of Pediatric Rheumatology, Department of Pediatrics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Maria Sylvia Vitalle
- Unit of Adolescence, Department of Pediatrics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Maria Isabel de Moraes-Pinto
- Research Laboratory, Division of Pediatric Infectious Diseases, Department of Pediatrics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Maria Teresa Terreri
- Unit of Pediatric Rheumatology, Department of Pediatrics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
| |
Collapse
|
14
|
Dirks J, Fischer J, Haase G, Holl-Wieden A, Hofmann C, Girschick H, Morbach H. CD21 lo/-CD27 -IgM - Double-Negative B Cells Accumulate in the Joints of Patients With Antinuclear Antibody-Positive Juvenile Idiopathic Arthritis. Front Pediatr 2021; 9:635815. [PMID: 33937147 PMCID: PMC8085394 DOI: 10.3389/fped.2021.635815] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/11/2021] [Indexed: 02/02/2023] Open
Abstract
Juvenile idiopathic arthritis (JIA) encompasses a heterogeneous group of diseases. The appearance of antinuclear antibodies (ANAs) in almost half of the patients suggests B cell dysregulation as a distinct pathomechanism in these patients. Additionally, ANAs were considered potential biomarkers encompassing a clinically homogenous subgroup of JIA patients. However, in ANA+ JIA patients, the site of dysregulated B cell activation as well as the B cell subsets involved in this process is still unknown. Hence, in this cross-sectional study, we aimed in an explorative approach at characterizing potential divergences in B cell differentiation in ANA+ JIA patients by assessing the distribution of peripheral blood (PB) and synovial fluid (SF) B cell subpopulations using flow cytometry. The frequency of transitional as well as switched-memory B cells was higher in PB of JIA patients than in healthy controls. There were no differences in the distribution of B cell subsets between ANA- and ANA+ patients in PB. However, the composition of SF B cells was different between ANA- and ANA+ patients with increased frequencies of CD21lo/-CD27-IgM- "double negative" (DN) B cells in the latter. DN B cells might be a characteristic subset expanding in the joints of ANA+ JIA patients and are potentially involved in the antinuclear immune response in these patients. The results of our explorative study might foster further research dissecting the pathogenesis of ANA+ JIA patients.
Collapse
Affiliation(s)
- Johannes Dirks
- Pediatric Immunology, University Children's Hospital, Würzburg, Germany
| | - Jonas Fischer
- Pediatric Immunology, University Children's Hospital, Würzburg, Germany
| | - Gabriele Haase
- Pediatric Immunology, University Children's Hospital, Würzburg, Germany
| | - Annette Holl-Wieden
- Pediatric Rheumatology and Osteology, University Children's Hospital, Würzburg, Germany
| | - Christine Hofmann
- Pediatric Rheumatology and Osteology, University Children's Hospital, Würzburg, Germany
| | | | - Henner Morbach
- Pediatric Immunology, University Children's Hospital, Würzburg, Germany.,Pediatric Rheumatology and Osteology, University Children's Hospital, Würzburg, Germany
| |
Collapse
|
15
|
Wennink RAW, Pandit A, Haasnoot AMJW, Hiddingh S, Kalinina Ayuso V, Wulffraat NM, Vastert BJ, Radstake TRDJ, de Boer JH, Kuiper JJW. Whole Transcriptome Analysis Reveals Heterogeneity in B Cell Memory Populations in Patients With Juvenile Idiopathic Arthritis-Associated Uveitis. Front Immunol 2020; 11:2170. [PMID: 33042130 PMCID: PMC7527539 DOI: 10.3389/fimmu.2020.02170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/10/2020] [Indexed: 12/25/2022] Open
Abstract
Purpose Patients with juvenile idiopathic arthritis (JIA) are prone to developing chronic anterior uveitis (JIA-U+). Although several risk factors for JIA-U+ have been identified, the underlying etiology is poorly understood. Histopathological studies demonstrate B cell infiltrates in eye tissues of patients with JIA-U+. Methods We performed transcriptome profiling of peripheral blood CD19-positive B cells taken from 14 cases with JIA-U+, 13 JIA cases without uveitis (JIA-U-), and five healthy controls. Deconvolution-based estimation was used to determine the immune cell fractions for each sample. Results Deconvolution results revealed that naive B cells made up on average 71% of the CD19-positive cell fractions analyzed. Differential expression analysis identified 614 differentially expressed genes (DEGs) between the groups at nominal significance and six genes at a false discovery rate of 5% (FDR < 0.05). Head-to-head comparison of all JIA-U- versus JIA-U+ revealed no DEGs in the CD19+ B cell pool (FDR < 0.05). However, principal component analysis based on a panel of key genes for B cell subsets revealed that JIA-U+ cases bifurcate into distinct clusters, characterized by markedly disparate expression for genes associated with specific memory B cell populations. CIBERSORT analysis of the overall transcriptome of the new uveitis cluster identified an increased proportion of memory B cells. Conclusion These data show that JIA-U- and JIA-U+ have a globally similar transcriptome considering the global peripheral CD19-positive B cell pool. However, heterogeneity in B cell memory genes among cases with uveitis suggests a role for specific memory B cell subsets in the etiology of JIA-U+.
Collapse
Affiliation(s)
- Roos A W Wennink
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Aridaman Pandit
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Anne-Mieke J W Haasnoot
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Sanne Hiddingh
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Viera Kalinina Ayuso
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Nico M Wulffraat
- Department of Pediatric Rheumatology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Bas J Vastert
- Department of Pediatric Rheumatology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Timothy R D J Radstake
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Joke H de Boer
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jonas J W Kuiper
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
16
|
Gatti A, Buccisano F, Scupoli MT, Brando B. The ISCCA flow protocol for the monitoring of anti-CD20 therapies in autoimmune disorders. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 100:194-205. [PMID: 32598578 DOI: 10.1002/cyto.b.21930] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/15/2020] [Accepted: 05/20/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Anti-CD20 monoclonals (MoAbs) are used in a variety of autoimmune disorders. The aim is to eliminate memory B cells sustaining the tissue damage and the production of pathogenic autoantibodies, while preserving naïve cells. The disappearance of memory B cells and the repopulation by naïve cells correlate with good clinical response, while the reappearance of memory B cells and plasmablasts correlates with relapse or resistance to therapy. Anti-CD20 induce extremely low B cell levels, requiring high-resolution techniques. The immune monitoring protocol developed by ISCCA is described and validated, to provide a standardized method for the clinical decision-making process during anti-CD20 therapies in autoimmune diseases. METHODS A 10-marker, 8-color staining panel (CD20-V450, CD45-V500c, CD4-FITC + sIgM-FITC, CD38-PE, CD3-PerCP Cy5.5, CD19-PE-Cy7, CD27-APC, CD8-APC H7 + sIgG-APC-H7) is used to identify B cells, plasma cells/blasts, naïve and memory B cells, sIgM+ and sIgG-switched memory B cells, T and NK cells, with high-sensitivity analysis (>106 CD45+ cells). RESULTS After an anti-CD20 dose, the B cell level is about zero in most patients. If B cells remain virtually absent (<0.1/μl), subsetting is not reliable nor meaningful. If B cells raise >0.3-0.5/μl, subsetting is possible and informative, acquiring >1.0-1.5 × 106 CD45+ events. Further testings can follow the quality of B cell repopulation. If B cells become detectable (>1/μl), the prevalence of memory B cells indicates non-responsiveness or a possible relapse. CONCLUSIONS The ISCCA Protocol is proposed for a standardized prospective monitoring of patients with autoimmune disorders, to assist the safe and rational usage of anti-CD20 therapies.
Collapse
Affiliation(s)
- Arianna Gatti
- Hematology Laboratory and Transfusion Center, Western Milan Area Hospital Consortium, Legnano, Milan, Italy
| | - Francesco Buccisano
- Department of Biomedicine and Prevention, Hematology, Tor Vergata University of Rome, Rome, Italy
| | - Maria T Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.,Research Center LURM (Interdepartmental Laboratory of Medical Research), University of Verona, Verona, Italy
| | - Bruno Brando
- Hematology Laboratory and Transfusion Center, Western Milan Area Hospital Consortium, Legnano, Milan, Italy
| |
Collapse
|
17
|
Fischer J, Dirks J, Haase G, Holl-Wieden A, Hofmann C, Girschick H, Morbach H. IL-21 + CD4 + T helper cells co-expressing IFN-γ and TNF-α accumulate in the joints of antinuclear antibody positive patients with juvenile idiopathic arthritis. Clin Immunol 2020; 217:108484. [PMID: 32485239 DOI: 10.1016/j.clim.2020.108484] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/16/2020] [Accepted: 05/26/2020] [Indexed: 12/23/2022]
Abstract
Juvenile Idiopathic Arthritis (JIA) is currently classified into seven subgroups. Recently, antinuclear antibody (ANA) positive JIA patients were suggested to encompass a clinically homogenous new subgroup. CD4+ T helper (Th) cells play an essential role in JIA pathogenesis. Herein, we analyzed cytokine expression in synovial fluid (SF) CD4+ Th cells of JIA patients by using flow cytometry and compared cytokine patterns between JIA subgroups. We could show increased frequencies of IL-21 expressing CD4+ Th cells in the joints of ANA+ Oligo-/Poly-JIA patients, which co-expressed the Th-1 cytokines IFN-γ/TNF-α. In contrast, frequencies of IL-17 expressing cells were lowest in the joints of ANA+ Oligo-/Poly-JIA but enriched in that of ERA-JIA patients. This is the first description of a diverse SF Th cell cytokine pattern in different JIA subgroups. Additionally, we could define IL-21 as an effector cytokine expressed in SF Th cell in a significant proportion of ANA+ JIA patients.
Collapse
Affiliation(s)
- Jonas Fischer
- Pediatric Immunology, , Department of Pediatrics, University of Würzburg, Germany
| | - Johannes Dirks
- Pediatric Immunology, , Department of Pediatrics, University of Würzburg, Germany
| | - Gabriele Haase
- Pediatric Immunology, , Department of Pediatrics, University of Würzburg, Germany
| | - Annette Holl-Wieden
- Pediatric Rheumatology, Department of Pediatrics, University of Würzburg, Germany
| | - Christine Hofmann
- Pediatric Rheumatology, Department of Pediatrics, University of Würzburg, Germany
| | | | - Henner Morbach
- Pediatric Immunology, , Department of Pediatrics, University of Würzburg, Germany; Pediatric Rheumatology, Department of Pediatrics, University of Würzburg, Germany.
| |
Collapse
|
18
|
Nagy A, Mosdosi B, Simon D, Dergez T, Berki T. Peripheral Blood Lymphocyte Analysis in Oligo- and Polyarticular Juvenile Idiopathic Arthritis Patients Receiving Methotrexate or Adalimumab Therapy: A Cross-Sectional Study. Front Pediatr 2020; 8:614354. [PMID: 33363071 PMCID: PMC7758242 DOI: 10.3389/fped.2020.614354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022] Open
Abstract
Juvenile idiopathic arthritis (JIA) is an umbrella term for seven distinct chronic immune-mediated diseases. Disease-modifying anti-rheumatic drugs (DMARD) are used to treat the underlying joint inflammation as well as extra-articular manifestations. Immunosuppression is a considerable side effect of the drugs. The main goal of this study was to investigate the effect of different JIA therapies on leukocyte subpopulations, which play a role in immune-defense. Three study groups were established. The first group consisted of JIA patients treated with methotrexate solely, the second one received a combination of methotrexate (MTX) and adalimumab (ADA). The control group was made up of the patients' healthy siblings. A total of 63 children were recruited. Fourty-one children with JIA and 22 healthy controls were included in the study. The absolute number of CD3+ T-cells was significantly elevated in patients treated with biological therapy compared to healthy controls (p2 = 0.017). In contrast, the number of CD56+ natural killer cells was significantly lower in children receiving biological therapy in comparison with healthy donors (p2 = 0.039). A significant alteration was also demonstrated between patients treated with MTX and MTX/ADA group concerning CD 19+ B-cells (p3 = 0.042). This is the first study that demonstrates significant alterations in the number of B-cells and T-cells with a relative decrease of NK-cell ratios in JIA patients receiving different DMARD therapy. Clinical Trial Registration: NCT03833271. 21.01.2019.
Collapse
Affiliation(s)
- Arnold Nagy
- Department of Paediatrics, University of Pecs, Medical School, Pecs, Hungary
| | - Bernadett Mosdosi
- Department of Paediatrics, University of Pecs, Medical School, Pecs, Hungary
| | - Diana Simon
- Department of Immunology and Biotechnology, University of Pecs, Medical School, Pecs, Hungary
| | - Timea Dergez
- Institute of Bioanalysis, University of Pecs, Medical School, Pecs, Hungary
| | - Timea Berki
- Department of Immunology and Biotechnology, University of Pecs, Medical School, Pecs, Hungary
| |
Collapse
|
19
|
Wilkinson MGL, Rosser EC. B Cells as a Therapeutic Target in Paediatric Rheumatic Disease. Front Immunol 2019; 10:214. [PMID: 30837988 PMCID: PMC6382733 DOI: 10.3389/fimmu.2019.00214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/24/2019] [Indexed: 12/12/2022] Open
Abstract
B cells carry out a central role in the pathogenesis of autoimmune disease. In addition to the production of autoantibodies, B cells can contribute to disease development by presenting autoantigens to autoreactive T cells and by secreting pro-inflammatory cytokines and chemokines which leads to the amplification of the inflammatory response. Targeting both the antibody-dependent and antibody-independent function of B cells in adult rheumatic disease has led to the advent of B cell targeted therapies in clinical practice. To date, whether B cell depletion could also be utilized for the treatment of pediatric disease is relatively under explored. In this review, we will discuss the role of B cells in the pathogenesis of the pediatric rheumatic diseases Juvenile Idiopathic Arthritis (JIA), Juvenile Systemic Lupus Erythematosus (JSLE) and Juvenile Dermatomyositis (JDM). We will also explore the rationale behind the use of B cell-targeted therapies in pediatric rheumatic disease by highlighting new case studies that points to their efficacy in JIA, JSLE, and JDM.
Collapse
Affiliation(s)
- Meredyth G Ll Wilkinson
- Infection, Immunity, Inflammation Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Arthritis Research UK Centre for Adolescent Rheumatology, University College London, UCLH and GOSH, London, United Kingdom.,NIHR Biomedical Research Centre, Great Ormond Street Hospital, London, United Kingdom
| | - Elizabeth C Rosser
- Infection, Immunity, Inflammation Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Arthritis Research UK Centre for Adolescent Rheumatology, University College London, UCLH and GOSH, London, United Kingdom.,NIHR Biomedical Research Centre, Great Ormond Street Hospital, London, United Kingdom
| |
Collapse
|
20
|
Zhao Q, Jung LK. Frequency of CD19 +CD24 hiCD38 hi regulatory B cells is decreased in peripheral blood and synovial fluid of patients with juvenile idiopathic arthritis: a preliminary study. Pediatr Rheumatol Online J 2018; 16:44. [PMID: 29973221 PMCID: PMC6033228 DOI: 10.1186/s12969-018-0262-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/20/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND To understand the relationship between regulatory B cells (Bregs) and juvenile idiopathic arthritis (JIA), we analyzed the percentages of Bregs and their function in peripheral blood (PB) and synovial fluid (SF) of JIA patients. METHODS Twenty-one JIA patients and 11 children with growing pain but without known rheumatic diseases as controls were included. The B cell phenotype and intracellular production of IL-10 of Bregs were assessed by flow cytometry. Mononuclear cells from PB and SF were stimulated to produce IL-10 in vitro for the identification of IL-10- producing regulatory B cells. RESULTS The percentage of CD24hiCD38hi Bregs in the PB of JIA patients was significantly decreased compared to that in controls, and it was even lower in the SF of JIA patients compared to that in the PB. CD24hiCD38hi Bregs frequency was significantly lower in the PB of RF-positive patients than in RF-negative patients. Frequency of IL-10-producing regulatory B cells (B10 cells) was significantly lower in active JIA patients than that in inactive patients. CONCLUSIONS The inability of the host to produce enough regulatory B cells in PB and especially in SF of JIA patients may contribute to the disease, especially the local inflammation.
Collapse
Affiliation(s)
- Qianzi Zhao
- grid.239560.bDivision of Rheumatology, Children’s National Medical Center, 111 Michigan Ave, NW, Washington, DC 20010 USA
| | - Lawrence K. Jung
- grid.239560.bDivision of Rheumatology, Children’s National Medical Center, 111 Michigan Ave, NW, Washington, DC 20010 USA
| |
Collapse
|
21
|
Aquilani A, Marafon DP, Marasco E, Nicolai R, Messia V, Perfetti F, Magni-Manzoni S, De Benedetti F. Predictors of Flare Following Etanercept Withdrawal in Patients with Rheumatoid Factor-negative Juvenile Idiopathic Arthritis Who Reached Remission while Taking Medication. J Rheumatol 2018; 45:956-961. [PMID: 29717035 DOI: 10.3899/jrheum.170794] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To evaluate the rate of flare after etanercept (ETN) withdrawal in patients with juvenile idiopathic arthritis (JIA) who attained clinical remission while taking medication, and to identify predictors of flare. METHODS Patients were included with oligo- (oJIA) and rheumatoid factor-negative polyarticular JIA (pJIA) who received a first course of ETN for at least 18 months, maintained clinically inactive disease (CID) for at least 6 months during treatment, and were followed for 12 months after ETN withdrawal. Demographic and clinical features were collected at onset, at baseline (initiation of ETN), and at time of disease flare. RESULTS After ETN withdrawal, 66 of the 110 patients enrolled (60%) flared with arthritis (of whom 7 flared with concurrent anterior uveitis; none with uveitis alone). The median time to flare was 4.3 months (interquartile range 2.5-6.4) with no evident differences between oJIA and pJIA. The number and type of joints involved at baseline and characteristics of ETN treatment/discontinuation were not associated with flare. Patients who flared were more frequently males (p = 0.034), positive for antinuclear antibody (ANA; p = 0.047), and had higher values of C-reactive protein (CRP; p = 0.012) at baseline. These variables remained significantly associated with flare in a multivariate logistic analysis, a model accounting for only 14% of the variability of the occurrence of the flare. CONCLUSION Our results show that a significant proportion of patients with JIA who maintain CID for at least 6 months experience a relapse after ETN withdrawal. Male sex, presence of ANA, and elevated CRP at baseline were associated with higher risk of flare.
Collapse
Affiliation(s)
- Angela Aquilani
- From the Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, Institute for Research and Health Care (IRCCS), Rome, Italy. .,A. Aquilani, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; D. Pires Marafon, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; E. Marasco, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; R. Nicolai, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; V. Messia, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. Perfetti, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; S. Magni-Manzoni, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. De Benedetti, MD, PhD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS.
| | - Denise Pires Marafon
- From the Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, Institute for Research and Health Care (IRCCS), Rome, Italy.,A. Aquilani, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; D. Pires Marafon, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; E. Marasco, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; R. Nicolai, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; V. Messia, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. Perfetti, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; S. Magni-Manzoni, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. De Benedetti, MD, PhD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS
| | - Emiliano Marasco
- From the Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, Institute for Research and Health Care (IRCCS), Rome, Italy.,A. Aquilani, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; D. Pires Marafon, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; E. Marasco, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; R. Nicolai, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; V. Messia, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. Perfetti, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; S. Magni-Manzoni, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. De Benedetti, MD, PhD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS
| | - Rebecca Nicolai
- From the Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, Institute for Research and Health Care (IRCCS), Rome, Italy.,A. Aquilani, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; D. Pires Marafon, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; E. Marasco, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; R. Nicolai, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; V. Messia, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. Perfetti, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; S. Magni-Manzoni, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. De Benedetti, MD, PhD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS
| | - Virginia Messia
- From the Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, Institute for Research and Health Care (IRCCS), Rome, Italy.,A. Aquilani, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; D. Pires Marafon, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; E. Marasco, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; R. Nicolai, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; V. Messia, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. Perfetti, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; S. Magni-Manzoni, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. De Benedetti, MD, PhD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS
| | - Francesca Perfetti
- From the Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, Institute for Research and Health Care (IRCCS), Rome, Italy.,A. Aquilani, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; D. Pires Marafon, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; E. Marasco, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; R. Nicolai, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; V. Messia, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. Perfetti, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; S. Magni-Manzoni, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. De Benedetti, MD, PhD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS
| | - Silvia Magni-Manzoni
- From the Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, Institute for Research and Health Care (IRCCS), Rome, Italy.,A. Aquilani, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; D. Pires Marafon, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; E. Marasco, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; R. Nicolai, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; V. Messia, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. Perfetti, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; S. Magni-Manzoni, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. De Benedetti, MD, PhD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS
| | - Fabrizio De Benedetti
- From the Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, Institute for Research and Health Care (IRCCS), Rome, Italy.,A. Aquilani, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; D. Pires Marafon, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; E. Marasco, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; R. Nicolai, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; V. Messia, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. Perfetti, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; S. Magni-Manzoni, MD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS; F. De Benedetti, MD, PhD, Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS
| |
Collapse
|