1
|
Dörner T, Bowman SJ, Fox R, Mariette X, Papas A, Grader-Beck T, Fisher BA, Barcelos F, De Vita S, Schulze-Koops H, Moots RJ, Junge G, Woznicki J, Sopala M, Avrameas A, Luo WL, Hueber W. Safety and Efficacy of Ianalumab in Patients With Sjögren's Disease: 52-Week Results From a Randomized, Placebo-Controlled, Phase 2b Dose-Ranging Study. Arthritis Rheumatol 2025; 77:560-570. [PMID: 39557617 DOI: 10.1002/art.43059] [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: 02/07/2024] [Revised: 09/16/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024]
Abstract
OBJECTIVE The objective of this study was to report 52-week safety and efficacy of ianalumab from phase 2b dose-finding study in patients with Sjögren's disease (SjD). METHODS Patients randomly received (1:1:1:1) ianalumab (5, 50, or 300 mg) or placebo subcutaneously every 4 weeks until week 24 (treatment period [TP]1). At week 24, patients on 300 mg were rerandomized to continue 300 mg or receive placebo until week 52 (TP2), patients on placebo were switched to ianalumab 150 mg, and patients on 5 and 50 mg directly entered posttreatment safety follow-up. Patients who discontinued treatment early or completed treatment entered safety follow-up (≥20 weeks). RESULTS During TP1, 190 patients were randomized (placebo = 49, 5 mg = 47, 50 mg = 47, 300 mg = 47). Of these 190 patients, 90 (47.4 %; 43 continued 300 mg and 47 received placebo) entered TP2, and 81 of 90 (90.0%) completed the study treatment. By week 52, efficacy was sustained in patients who continued 300 mg in TP2 (EULAR Sjögren's Syndrome Disease Activity Index, EULAR Sjögren's Syndrome Patient Reported Index, patient global assessment, and physician global assessment change from week 24: -1.45, -0.46, -4.69, and -6.86, respectively). Stimulated salivary flow rates and autoantibody levels numerically improved in the 300 mg group. Treatment-emergent adverse events were not dose-dependent, except for injection-site reactions. Cases of decreased neutrophil counts (Common Terminology Criteria for Adverse Events v4.03 grade 3 according to laboratory listings) were observed in three patients during the posttreatment follow-up, occurring at 3.5, 5.5, and 3 months, after the last ianalumab administration. None were associated with infection except one incidental finding of asymptomatic cytomegalovirus infection (IgM-positive). CONCLUSION In patients with SjD, ianalumab 300 mg demonstrated sustained efficacy through week 52 and a favorable safety profile up to two years of follow-up.
Collapse
Affiliation(s)
| | - Simon J Bowman
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Robert Fox
- Scripps Memorial Hospital and Research Institute, La Jolla, California
| | - Xavier Mariette
- Université Paris-Saclay, Paris, France, and Hôpital Bicêtre, AP-HP, Le Kremlin Bicêtre, France
| | - Athena Papas
- Tufts School of Dental Medicine, Boston, Massachusetts
| | | | - Benjamin A Fisher
- University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, and NIHR Birmingham Biomedical Research Centre, Birmingham, United Kingdom
| | | | | | | | - Robert J Moots
- Aintree University Hospital, Liverpool, United Kingdom, and Edge Hill University, Ormskirk, United Kingdom
| | | | - Janice Woznicki
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | | | | | - Wen-Lin Luo
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | | |
Collapse
|
2
|
Berthe J, Poudel P, Segerer FJ, Jennings EC, Ng F, Surace M, Andoni A, Testori M, Saraiya M, Vuko M, Hessel H, Heininen-Brown M, Blando J, Jones EV, Willis SE, Galon J, van de Ven R, de Gruijl TD, Angell HK. Exploring the impact of tertiary lymphoid structures maturity in NSCLC: insights from TLS scoring. Front Immunol 2024; 15:1422206. [PMID: 39376565 PMCID: PMC11457083 DOI: 10.3389/fimmu.2024.1422206] [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: 04/23/2024] [Accepted: 08/26/2024] [Indexed: 10/09/2024] Open
Abstract
Tertiary Lymphoid Structures (TLS) are lymphoid structures commonly associated with improved survival of cancer patients and response to immunotherapies. However, conflicting reports underscore the need to consider TLS heterogeneity and multiple features such as TLS size, composition, and maturation status, when assessing their functional impact. With the aim of gaining insights into TLS biology and evaluating the prognostic impact of TLS maturity in Non-Small Cell Lung Carcinoma (NSCLC), we developed a multiplex immunofluorescent (mIF) panel including T cell (CD3, CD8), B cell (CD20), Follicular Dendritic cell (FDC) (CD21, CD23) and mature dendritic cell (DC-LAMP) markers. We deployed this panel across a cohort of primary tumor resections from NSCLC patients (N=406) and established a mIF image analysis workstream to specifically detect TLS structures and evaluate the density of each cell phenotype. We assessed the prognostic significance of TLS size, number, and composition, to develop a TLS scoring system representative of TLS biology within a tumor. TLS relative area, (total TLS area divided by the total tumor area), was the most prognostic TLS feature (C-index: 0.54, p = 0.04). CD21 positivity was a marker driving the favorable prognostic impact, where CD21+ CD23- B cells (C-index: 0.57, p = 0.04) and CD21+ CD23- FDC (C-index: 0.58, p = 0.01) were the only prognostic cell phenotypes in TLS. Combining the three most robust prognostic TLS features: TLS relative area, the density of B cells, and FDC CD21+ CD23- we generated a TLS scoring system that demonstrated strong prognostic value in NSCLC when considering the effect of age, sex, histology, and smoking status. This TLS Score also demonstrated significant association with Immunoscore, EGFR mutational status and gene expression-based B-cell and TLS signature scores. It was not correlated with PD-L1 status in tumor cells or immune cells. In conclusion, we generated a prognostic TLS Score representative of the TLS heterogeneity and maturity undergoing within NSCLC tissues. This score could be used as a tool to explore how TLS presence and maturity impact the organization of the tumor microenvironment and support the discovery of spatial biomarker surrogates of TLS maturity, that could be used in the clinic.
Collapse
Affiliation(s)
- Julie Berthe
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Pawan Poudel
- Oncology Data Science, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Felix J. Segerer
- Computational Pathology, Oncology R&D, AstraZeneca, Munich, Germany
| | - Emily C. Jennings
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, United States
| | - Felicia Ng
- Oncology Data Science, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Michael Surace
- Translational Medicine, Oncology R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Alma Andoni
- Translational Medicine, Oncology R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Marco Testori
- Computational Pathology, Oncology R&D, AstraZeneca, Munich, Germany
| | - Megha Saraiya
- Computational Pathology, Oncology R&D, AstraZeneca, Munich, Germany
| | - Miljenka Vuko
- Computational Pathology, Oncology R&D, AstraZeneca, Munich, Germany
| | - Harald Hessel
- Computational Pathology, Oncology R&D, AstraZeneca, Munich, Germany
| | | | - Jorge Blando
- Translational Medicine, Oncology R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Emma V. Jones
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Sophie E. Willis
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Sorbonne Université, Université Paris Cité, Centre de Recherche des Cordeliers, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Rieneke van de Ven
- Department of Otolaryngology, Head and Neck Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology Theme, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, Netherlands
| | - Tanja D. de Gruijl
- Cancer Center Amsterdam, Cancer Biology and Immunology Theme, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, Netherlands
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Helen K. Angell
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
3
|
Mostkowska A, Rousseau G, Raynal NJM. Repurposing of rituximab biosimilars to treat B cell mediated autoimmune diseases. FASEB J 2024; 38:e23536. [PMID: 38470360 DOI: 10.1096/fj.202302259rr] [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: 11/01/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/13/2024]
Abstract
Rituximab, the first monoclonal antibody approved for the treatment of lymphoma, eventually became one of the most popular and versatile drugs ever in terms of clinical application and revenue. Since its patent expiration, and consequently, the loss of exclusivity of the original biologic, its repurposing as an off-label drug has increased dramatically, propelled by the development and commercialization of its many biosimilars. Currently, rituximab is prescribed worldwide to treat a vast range of autoimmune diseases mediated by B cells. Here, we present a comprehensive overview of rituximab repurposing in 115 autoimmune diseases across 17 medical specialties, sourced from over 1530 publications. Our work highlights the extent of its off-label use and clinical benefits, underlining the success of rituximab repurposing for both common and orphan immune-related diseases. We discuss the scientific mechanism associated with its clinical efficacy and provide additional indications for which rituximab could be investigated. Our study presents rituximab as a flagship example of drug repurposing owing to its central role in targeting cluster of differentiate 20 positive (CD20) B cells in 115 autoimmune diseases.
Collapse
Affiliation(s)
- Agata Mostkowska
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Guy Rousseau
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Noël J-M Raynal
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Centre de recherche du CHU Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
4
|
Berti A, Hillion S, Konig MF, Moura MC, Hummel AM, Carmona E, Peikert T, Fervenza FC, Kallenberg CGM, Langford CA, Merkel PA, Monach PA, Seo P, Spiera RF, Brunetta P, Clair EW, Harris KM, Stone JH, Grandi G, Pers JO, Specks U, Cornec D. Autoreactive Plasmablasts After B Cell Depletion With Rituximab and Relapses in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Arthritis Rheumatol 2023; 75:736-747. [PMID: 36281741 PMCID: PMC10280646 DOI: 10.1002/art.42388] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/04/2022] [Accepted: 10/11/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Autoreactive B cells are responsible for antineutrophil cytoplasmic antibody (ANCA) production in ANCA-associated vasculitis (AAV). Rituximab (RTX) depletes circulating B cells, including autoreactive B cells. We aimed to evaluate changes and associations with relapse of the circulating autoreactive B cell pool following therapeutic B cell depletion in AAV. METHODS Sequential flow cytometry was performed on 148 samples of peripheral blood mononuclear cells from 23 patients with proteinase 3 (PR3)-ANCA-positive AAV who were treated with RTX for remission induction and monitored after stopping therapy during long-term follow-up in a prospective clinical trial. PR3 was used as a ligand to target autoreactive PR3-specific (PR3+) B cells. B cell recurrence was considered as the first blood sample with ≥10 B cells/μl after RTX treatment. RESULTS At B cell recurrence, PR3+ B cell frequency among B cells was higher than baseline (P < 0.01). Within both PR3+ and total B cells, frequencies of transitional and naive subsets were higher at B cell recurrence than at baseline, while memory subsets were lower (P < 0.001 for all comparisons). At B cell recurrence, frequencies of B cells and subsets did not differ between patients who experienced relapse and patients who remained in remission. In contrast, the plasmablast frequency within the PR3+ B cell pool was higher in patients who experienced relapse and associated with a shorter time to relapse. Frequencies of PR3+ plasmablasts higher than baseline were more likely to be found in patients who experienced relapse within the following 12 months compared to those in sustained remission (P < 0.05). CONCLUSION The composition of the autoreactive B cell pool varies significantly following RTX treatment in AAV, and early plasmablast enrichment within the autoreactive pool is associated with future relapses.
Collapse
Affiliation(s)
- Alvise Berti
- Division of Pulmonary & Critical Care Medicine, Thoracic Disease Research Unit, Mayo Clinic, Rochester, Minnesota, and Center for Medical Sciences (CISMed), Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Italy, and Rheumatology Unit, Santa Chiara Hospital, APSS Trento, Italy
| | - Sophie Hillion
- Jacques-Olivier Pers, DDS, PhD, Divi Cornec, MD, PhD: Université de Bretagne Occidendale, Brest, Bretagne, France
| | - Maximilian F. Konig
- Division of Rheumatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marta Casal Moura
- Division of Pulmonary & Critical Care Medicine, Thoracic Disease Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Amber M. Hummel
- Division of Pulmonary & Critical Care Medicine, Thoracic Disease Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Eva Carmona
- Division of Pulmonary & Critical Care Medicine, Thoracic Disease Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Tobias Peikert
- Division of Pulmonary & Critical Care Medicine, Thoracic Disease Research Unit, Mayo Clinic, Rochester, Minnesota
| | | | - Cees G. M. Kallenberg
- Department of Rheumatology and Clinical Immunology, University of Groningen, Groningen, The Netherlands
| | | | - Peter A. Merkel
- Division of Rheumatology, Department of Medicine, and Department of Biostatistics, Epidemiology, and Informatics, Division of Clinical Epidemiology, University of Pennsylvania, Philadelphia
| | | | - Philip Seo
- Division of Rheumatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert F. Spiera
- Weill Cornell Medical College, Hospital for Special Surgery, New York
| | | | | | | | - John H. Stone
- Massachusetts General Hospital Rheumatology Unit, Boston
| | - Guido Grandi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Italy
| | - Jacques-Olivier Pers
- Jacques-Olivier Pers, DDS, PhD, Divi Cornec, MD, PhD: Université de Bretagne Occidendale, Brest, Bretagne, France
| | - Ulrich Specks
- Division of Pulmonary & Critical Care Medicine, Thoracic Disease Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Divi Cornec
- Jacques-Olivier Pers, DDS, PhD, Divi Cornec, MD, PhD: Université de Bretagne Occidendale, Brest, Bretagne, France
| |
Collapse
|
5
|
Comprehensive overview of autoantibody isotype and subclass distribution. J Allergy Clin Immunol 2022; 150:999-1010. [DOI: 10.1016/j.jaci.2022.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/27/2022] [Accepted: 05/13/2022] [Indexed: 11/06/2022]
|
6
|
Zhou B, Zhang H, Su X, Luo Y, Li X, Yu C, Xie Q, Xia X, He G, Yang L. Therapeutic effects of a novel BAFF blocker on arthritis. Signal Transduct Target Ther 2019; 4:19. [PMID: 31231554 PMCID: PMC6565627 DOI: 10.1038/s41392-019-0051-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/01/2019] [Accepted: 04/10/2019] [Indexed: 02/05/2023] Open
Abstract
B-cell targeted therapy is effective for autoimmune diseases such as systemic lupus erythematosus and rheumatoid arthritis (RA), although there are setbacks in RA clinical trials. In this study, we designed a novel B-cell activating factor (BAFF) antagonist: BAFF-Trap, a recombinant glycoprotein with BAFF-binding domains of two BAFF receptors (TACI and Br3) linked to Fc domain of human IgG1. Unlike TACI-Fc, BAFF-Trap bound BAFF but not APRIL (a proliferation-inducing ligand), and significantly suppressed the development of collagen-induced arthritis and adjuvant-induced arthritis. Furthermore, BAFF-Trap inhibited proinflammatory cytokine expression, ameliorated joint damage and suppressed B- and T-cell activation. BAFF-Trap reduced dendritic cells in joints, and increased regulatory T cell, regulatory B-cell, and M2 macrophage. The function of BAFF-Trap was related to inhibition of canonical and noncanonical NF-κB activation. Thus, BAFF-Trap may be a valuable agent for the effective treatment of RA.
Collapse
Affiliation(s)
- Bailing Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan China
| | - Hailong Zhang
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan China
- Henan Engineering Laboratory of Antibody Medicine, Henan International United Laboratory of Antibody Medicine, Key laboratory of Cellular and Molecular Immunology, College of Medicine, Henan University, Kaifeng, Henan China
| | - Xiaoqing Su
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan China
| | - Yi Luo
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan China
| | - Xiaopeng Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan China
| | - Chaoheng Yu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan China
| | - Qibing Xie
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan China
| | - Xuyang Xia
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan China
| | - Gu He
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan China
| | - Li Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan China
| |
Collapse
|
7
|
Standardisation of flow cytometry for whole blood immunophenotyping of islet transplant and transplant clinical trial recipients. PLoS One 2019; 14:e0217163. [PMID: 31116766 PMCID: PMC6530858 DOI: 10.1371/journal.pone.0217163] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/06/2019] [Indexed: 01/03/2023] Open
Abstract
Understanding the immunological phenotype of transplant recipients is important to improve outcomes and develop new therapies. Immunophenotyping of whole peripheral blood (WPB) by flow cytometry is a rapid method to obtain large amounts of data relating to the outcomes of different transplant treatments with limited patient impact. Healthy individuals and patients with type 1 diabetes (T1D) enrolled in islet transplantation were recruited and WPB was collected. 46 fluorochrome-conjugated mouse-anti-human antibodies were used (43 of 46 antibodies were titrated). BD cytometer setup and tracking beads were used to characterize and adjust for cytometer performance. Antibody cocktails were pre-mixed <60 minutes before staining. Multicolour panels were designed based on fluorochrome brightness, antigen density, co-expression, and fluorochrome spillover into non-primary detectors in each panel on a 5 laser flow cytometer. WPB sample staining used 50–300 μl WPB for each panel and was performed within 2 hours of blood sample collection. Samples were acquired on a BD-LSRFortessa. The operating procedures, including specimen collection, antibody cocktails, staining protocol, flow-cytometer setup and data analysis, were standardized. The staining index of 43 antibodies and the spillover spreading matrix for each panel was calculated. The final concentrations for the 46 antibodies used was determined for staining of WPB samples. Absolute cell-count and 7 leukocyte profiling panels consisting of subsets and/or status of granulocytes, monocytes, dendritic, B, NK, and T cells including regulatory T cells (Tregs) and NKT were designed and established on a 5 laser BD-LSR Fortessa. 13 T1D patients, including 4 islet transplant recipients and 8 healthy controls, were evaluated. The ability to reproducibly measure immune subsets and immune-profiles of islet transplant patients up to 18 months post transplantation has been established as a tool to measure immune cell reconstitution after transplantation.
Collapse
|
8
|
Yu CG, Bondada V, Ghoshal S, Singh R, Pistilli CK, Dayaram K, Iqbal H, Sands M, Davis KL, Bondada S, Geddes JW. Repositioning Flubendazole for Spinal Cord Injury. J Neurotrauma 2019; 36:2618-2630. [PMID: 30747048 DOI: 10.1089/neu.2018.6160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
We previously reported the serendipitous observation that fenbendazole, a benzimidazole anthelmintic, improved functional and pathological outcomes following thoracic spinal cord contusion injury in mice when administered pre-injury. Fenbendazole is widely used in veterinary medicine. However, it is not approved for human use and it was uncertain if only post-injury administration would offer similar benefits. In the present study we evaluated post-injury administration of a closely related, human anthelmintic drug, flubendazole, using a rat spinal cord contusion injury model. Flubendazole, administered i.p. 5 or 10 mg/kg day, beginning 3 h post-injury and daily thereafter for 2 or 4 weeks, resulted in improved locomotor function after contusion spinal cord injury (SCI) compared with vehicle-treated controls. Histological analysis of spinal cord sections showed that such treatment with flubendazole also reduced lesion volume and improved total tissue sparing, white matter sparing, and gray matter sparing. Flubendazole inhibited the activation of glial fibrillary acidic protein (GFAP); suppressed cyclin B1 expression and Bruton tyrosine kinase activation, markers of B cell activation/proliferation and inflammation; and reduced B cell autoimmune response. Together, these results suggest the use of the benzimidazole anthelmintic flubendazole as a potential therapeutic for SCI.
Collapse
Affiliation(s)
- Chen Guang Yu
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Vimala Bondada
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Sarbani Ghoshal
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Ranjana Singh
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Christina K Pistilli
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Kavi Dayaram
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Hina Iqbal
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Madison Sands
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Kate L Davis
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Subarrao Bondada
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky
| | - James W Geddes
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
9
|
Crayne CB, Eloseily E, Mannion ML, Azerf SP, Weiser P, Beukelman T, Stoll ML, Feig DI, Prescott Atkinson T, Cron RQ. Rituximab treatment for chronic steroid-dependent Henoch-Schonlein purpura: 8 cases and a review of the literature. Pediatr Rheumatol Online J 2018; 16:71. [PMID: 30428889 PMCID: PMC6236882 DOI: 10.1186/s12969-018-0285-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/25/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Henoch-Schonlein purpura (HSP) is a small vessel vasculitis that is characterized by non-thrombocytopenic purpura, abdominal pain, arthritis, and glomerulonephritis. Typically, HSP is self-limited requiring only supportive care, but more severe cases may require corticosteroid (CS) treatment. Rarely, a subset of these patients has persistent rash, arthritis, abdominal involvement, or renal disease despite treatment with CS, or has disease recurrence on CS tapering. Refractory HSP has been effectively treated with a variety of CS sparing therapies. For life-threatening refractory HSP, the B cell depleting agent, rituximab (RTX), has been reported as beneficial for children with substantial renal or central nervous system involvement. However, RTX use for children with less severe HSP, but chronic CS dependent disease refractory to CS sparing immunomodulatory agents, has been less well explored. Herein, we describe 8 children treated with RTX for chronic refractory HSP and report a reduction in recurrent hospitalizations and eventual CS discontinuation. METHODS This is a retrospective analysis of eight children who were treated with RTX for chronic CS dependent HSP during the years 2006-2014 at a single institution. A chart review of the electronic medical record was performed to determine the presenting symptoms, the type and duration of treatment received, and the number of hospitalizations prior to and after RTX. The number of hospitalizations and oral corticosteroid burden were analyzed using the Wilcoxon signed rank test. RESULTS Prior to receiving RTX, seven patients had at least one hospitalization for HSP (median 1.5, range 0-3). Following RTX, only two patients were hospitalized, each a single time for recurrent abdominal pain. The median oral CS burden was 0.345 mg/kg/day before RTX and 0 mg/kg/day at 6 months (p = 0.078), 1 year (p = 0.0625), and 2 years (p = 0.03) following RTX infusion. Seven out of eight children met remission criteria, defined as no active rash, arthritis, nephritis (hematuria and proteinuria), or gastrointestinal distress following RTX. No serious adverse events were noted. CONCLUSION Overall, RTX effectively reduced the number of hospital admissions and oral CS burden. RTX also helped most all children achieve clinical remission. RTX appears to be an effective and safe alternative for chronic CS dependent and immunomodulatory refractory childhood HSP.
Collapse
Affiliation(s)
- Courtney B. Crayne
- 0000000106344187grid.265892.2Department of Pediatrics, Division of Rheumatology, University of Alabama at Birmingham, 1600 7th Ave S, CPPN G10, Birmingham, AL 35233 USA
| | - Esraa Eloseily
- 0000000106344187grid.265892.2Department of Pediatrics, Division of Rheumatology, University of Alabama at Birmingham, 1600 7th Ave S, CPPN G10, Birmingham, AL 35233 USA
| | - Melissa L. Mannion
- 0000000106344187grid.265892.2Department of Pediatrics, Division of Rheumatology, University of Alabama at Birmingham, 1600 7th Ave S, CPPN G10, Birmingham, AL 35233 USA
| | - Saji P. Azerf
- 0000000106344187grid.265892.2School of Medicine, University of Alabama at Birmingham, 510 20th St S, #12, Birmingham, AL 35233 USA
| | - Peter Weiser
- 0000000106344187grid.265892.2Department of Pediatrics, Division of Rheumatology, University of Alabama at Birmingham, 1600 7th Ave S, CPPN G10, Birmingham, AL 35233 USA
| | - Timothy Beukelman
- 0000000106344187grid.265892.2Department of Pediatrics, Division of Rheumatology, University of Alabama at Birmingham, 1600 7th Ave S, CPPN G10, Birmingham, AL 35233 USA
| | - Matthew L. Stoll
- 0000000106344187grid.265892.2Department of Pediatrics, Division of Rheumatology, University of Alabama at Birmingham, 1600 7th Ave S, CPPN G10, Birmingham, AL 35233 USA
| | - Daniel I. Feig
- 0000000106344187grid.265892.2Division of Nephrology, Department of Pediatrics, University of Alabama at Birmingham, 1600 7th Ave S, ACC 516, Birmingham, AL 35233 USA
| | - T. Prescott Atkinson
- 0000000106344187grid.265892.2Division of Allergy & Immunology, Department of Pediatrics, University of Alabama at Birmingham, 1600 7th Ave S, CPPN M20, Birmingham, AL 35233 USA
| | - Randy Quentin Cron
- Department of Pediatrics, Division of Rheumatology, University of Alabama at Birmingham, 1600 7th Ave S, CPPN G10, Birmingham, AL, 35233, USA.
| |
Collapse
|
10
|
Teplyakov A, Obmolova G, Luo J, Gilliland GL. Crystal structure of B-cell co-receptor CD19 in complex with antibody B43 reveals an unexpected fold. Proteins 2018; 86:495-500. [PMID: 29490423 DOI: 10.1002/prot.25485] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 02/11/2018] [Accepted: 02/24/2018] [Indexed: 12/27/2022]
Abstract
CD19 is a transmembrane protein expressed on malignant B cells, but not in other lineages or other tissues, which makes it an attractive target for monoclonal antibody-mediated immunotherapy. Anti-CD19 antibody B43 was utilized in a bispecific T-cell engager (BiTE) blinatumomab that demonstrated potency for the treatment of relapsed acute lymphoblastic leukemia. To gain insight into the mechanism of action of the antibody, the crystal structure of B43 Fab was determined in complex with CD19 and in the unbound form. The structure revealed the binding epitope, explained the lack of cross-reactivity toward non-human species, and suggested the key-and-lock mechanism of antigen recognition. Most unexpectedly, the structure revealed a unique molecular topology of CD19. Rather than a tandem of c-type immunoglobulin folds predicted from the amino acid sequence, the extracellular domain of CD19 exhibits an elongated β-sandwich formed by two immunoglobulin folds by swapping their C-terminal halves. This is the first structure of CD19, which has no sequence homologs.
Collapse
Affiliation(s)
- Alexey Teplyakov
- Janssen Research and Development, LLC, 1400 McKean Road, Spring House, Pennsylvania, 19477
| | - Galina Obmolova
- Janssen Research and Development, LLC, 1400 McKean Road, Spring House, Pennsylvania, 19477
| | - Jinquan Luo
- Janssen Research and Development, LLC, 1400 McKean Road, Spring House, Pennsylvania, 19477
| | - Gary L Gilliland
- Janssen Research and Development, LLC, 1400 McKean Road, Spring House, Pennsylvania, 19477
| |
Collapse
|
11
|
Kumar S, Ward BR, Irani AM. Future Prospects of Biologic Therapies for Immunologic Diseases. Immunol Allergy Clin North Am 2017; 37:431-448. [PMID: 28366486 DOI: 10.1016/j.iac.2017.01.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This article presents an overview of future uses for biologic therapies in the treatment of immunologic and allergic conditions. Discussion is centered on the use of existing therapies outside of their current indication or on new therapies that are close to approval. This information may help familiarize practicing allergists and immunologists with therapies they may soon encounter in their practice as well as help identify conditions and treatments that will require further study in the near future.
Collapse
Affiliation(s)
- Santhosh Kumar
- Division of Allergy and Immunology, Children's Hospital of Richmond, Virginia Commonwealth University, CHoR Pavilion, 5th Floor, 1000 East Broad Street, Richmond, VA 23298-0225, USA.
| | - Brant R Ward
- Division of Allergy and Immunology, Children's Hospital of Richmond, Virginia Commonwealth University, CHoR Pavilion, 5th Floor, 1000 East Broad Street, Richmond, VA 23298-0225, USA; Division of Rheumatology, Allergy, and Immunology, Virginia Commonwealth University, McGuire Hall, Room 4-115A, 1112 East Clay Street, Richmond, VA 23298-0263, USA; Department of Microbiology and Immunology, Virginia Commonwealth University, 1101 East Marshall Street, P.O. Box 980678, Richmond, VA 23298, USA
| | - Anne-Marie Irani
- Division of Allergy and Immunology, Children's Hospital of Richmond, Virginia Commonwealth University, CHoR Pavilion, 5th Floor, 1000 East Broad Street, Richmond, VA 23298-0225, USA
| |
Collapse
|
12
|
Nguyen A, Gresle M, Marshall T, Butzkueven H, Field J. Monoclonal antibodies in the treatment of multiple sclerosis: emergence of B-cell-targeted therapies. Br J Pharmacol 2017; 174:1895-1907. [PMID: 28319650 PMCID: PMC5466523 DOI: 10.1111/bph.13780] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/26/2017] [Accepted: 03/03/2017] [Indexed: 12/16/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the CNS, and one of the most common causes of disability in young adults. Over the last decade, new disease-modifying therapies have emerged, including monoclonal antibodies (mAbs) that provide highly targeted therapies with greater efficacy than platform therapies. In particular, monoclonal antibodies directed against CD20-positive B cells have shown remarkable results in recent clinical trials and renewed interest in the mechanism of B cell-depleting therapies to ameliorate relapse activity and progression in MS. Here, we review the mechanisms of action and clinical evidence of approved and emerging mAbs, with a focus on B cell-targeted therapies.
Collapse
Affiliation(s)
- Ai‐Lan Nguyen
- Melbourne Brain Centre and Department of Medicine at the Royal Melbourne HospitalUniversity of MelbourneParkvilleVic.Australia
| | - Melissa Gresle
- Melbourne Brain Centre and Department of Medicine at the Royal Melbourne HospitalUniversity of MelbourneParkvilleVic.Australia
| | - Tessa Marshall
- Multiple Sclerosis DivisionThe Florey Institute of Neuroscience and Mental HealthParkvilleVic.Australia
| | - Helmut Butzkueven
- Melbourne Brain Centre and Department of Medicine at the Royal Melbourne HospitalUniversity of MelbourneParkvilleVic.Australia
- Eastern HealthMonash UniversityClaytonVic.Australia
| | - Judith Field
- Multiple Sclerosis DivisionThe Florey Institute of Neuroscience and Mental HealthParkvilleVic.Australia
- Department of Anatomy and NeuroscienceUniversity of MelbourneParkvilleVic.Australia
| |
Collapse
|
13
|
Zhao C, Li HZ, Zhao DD, Ma C, Wu F, Bai YN, Zhang M, Li ZY, Guo J. Increased Circulating T Follicular Helper Cells Are Inhibited by Rituximab in Neuromyelitis Optica Spectrum Disorder. Front Neurol 2017; 8:104. [PMID: 28360886 PMCID: PMC5350120 DOI: 10.3389/fneur.2017.00104] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 03/02/2017] [Indexed: 12/12/2022] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is a severe autoimmune disease of the central nervous system. The existence of autoantibody targeting aquaporin-4 (AQP4-Ab) indicates the involvement of humoral immunity in the pathogenesis of this disease. Rituximab (RTX), a monoclonal antibody against CD20, has been used to treat NMOSD by depleting circulating B cells and overall satisfactory outcome has been achieved. Although T follicular helper cells have been proved to regulate B cell activation and antibody production, the role of these cells in NMOSD and the impact of RTX treatment on these cells remain less understood. In this study, we found that frequencies of circulating T follicular helper (cTfh) cells and B cells together with the related cytokines, IL-21 and IL-6, were closely correlated with disease activity of NMOSD. Furthermore, B cell depletion with RTX treatment inhibited the expansion of cTfh cells, and these effects were achieved through eliminating IL-6-producing B cells and blocking the direct contact between cTfh cells and B cells. These findings imply the complicated cross talk between cTfh cells and B cells and may provide a novel therapeutic target for NMOSD.
Collapse
Affiliation(s)
- Cong Zhao
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| | - Hong-Zeng Li
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| | - Dai-Di Zhao
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| | - Chao Ma
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| | - Fang Wu
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China; Department of Neurology, Xi'an Children's Hospital, Xi'an, China
| | - Ya-Nan Bai
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| | - Min Zhang
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| | - Zhu-Yi Li
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| | - Jun Guo
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University , Xi'an , China
| |
Collapse
|
14
|
B Lymphocytes in Multiple Sclerosis: Bregs and BTLA/CD272 Expressing-CD19+ Lymphocytes Modulate Disease Severity. Sci Rep 2016; 6:29699. [PMID: 27412504 PMCID: PMC4944189 DOI: 10.1038/srep29699] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 06/23/2016] [Indexed: 01/18/2023] Open
Abstract
B lymphocytes contribute to the pathogenesis of Multiple Sclerosis (MS) by secreting antibodies and producing cytokines. This latter function was analyzed in myelin olygodendrocyte protein (MOG)-stimulated CD19+ B lymphocytes of 71 MS patients with different disease phenotypes and 40 age-and sex-matched healthy controls (HC). Results showed that: 1) CD19+/TNFα+, CD19+/IL-12+ and CD19+/IFNγ+ lymphocytes are significantly increased in primary progressive (PP) compared to secondary progressive (SP), relapsing-remitting (RR), benign (BE) MS and HC; 2) CD19+/IL-6+ lymphocytes are significantly increased in PP, SP and RR compared to BEMS and HC; and 3) CD19+/IL-13+, CD19+/IL-10+, and CD19+/IL-10+/TGFβ+ (Bregs) B lymphocytes are reduced overall in MS patients compared to HC. B cells expressing BTLA, a receptor whose binding to HVEM inhibits TcR-initiated cytokine production, as well as CD19+/BTLA+/IL-10+ cells were also significantly overall reduced in MS patients compared to HC. Analyses performed in RRMS showed that fingolimod-induced disease remission is associated with a significant increase in Bregs, CD19+/BTLA+, and CD19+/BTLA+/IL-10+ B lymphocytes. B lymphocytes participate to the pathogenesis of MS via the secretion of functionally-diverse cytokines that might play a role in determining disease phenotypes. The impairment of Bregs and CD19+/BTLA+ cells, in particular, could play an important pathogenic role in MS.
Collapse
|
15
|
Störch H, Zimmermann B, Resch B, Tykocinski LO, Moradi B, Horn P, Kaya Z, Blank N, Rehart S, Thomsen M, Lorenz HM, Neumann E, Tretter T. Activated human B cells induce inflammatory fibroblasts with cartilage-destructive properties and become functionally suppressed in return. Ann Rheum Dis 2016; 75:924-32. [PMID: 25985971 DOI: 10.1136/annrheumdis-2014-206965] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/26/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cross-talk between synovial fibroblasts (SF) and immune cells is suggested to play a crucial role in inflammation and chronification of rheumatoid arthritis (RA). The contribution of B cells in this process is poorly defined. METHODS Here, primary B cells from healthy donors were polyclonally activated and cocultured with SF of non-synovitic origin from patients with osteoarthritis. RESULTS In B-SF cocultures the concentrations of interleukin 6 (IL-6) and IL-8 increased manifold compared with single cultures even under physical separation and remained stable for several days after B-cell removal. Intracellular staining confirmed SF as key producers of IL-6 and IL-8, and B cells as main producers of tumour necrosis factor alpha (TNFα) and IL-1ß. Blocking experiments with a combination of anti-TNFα-antibodies and rIL-1RA significantly reduced SF cytokine production by up to 90%, suggesting that B-cell-derived TNFα and IL-1ß were crucial mediators of SF activation. Interestingly, B-cell cytokine production, CD25 expression and proliferation decreased in cocultures by at least 50%, demonstrating a negative regulatory loop towards the activated B cells. Inhibition of activin receptor-like kinase 5, a crucial component of the tumour growth factor ß (TGFß) signalling pathway, partly restored B-cell proliferation, suggesting a contribution of SF-derived TGFß in B-cell suppression. Besides cytokines, B-cell-activated SF also upregulated secretion of matrix metalloproteases such as MMP-3, thereby acquiring potential tissue destructive properties. This was confirmed by their invasion into human cartilage in the severe combined immunodeficiency mouse fibroblast invasion model in vivo. CONCLUSIONS Interaction with activated B cells leads to conversion of non-arthritic SF into SF with a proinflammatory and aggressive RA-like phenotype, thereby suggesting a new, so far unrecognised role for B cells in RA pathogenesis.
Collapse
Affiliation(s)
- Hannah Störch
- Division of Rheumatology, Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Birgit Zimmermann
- Department of Internal Medicine and Rheumatology, University of Giessen, Kerckhoff-Klinik, Bad Nauheim, Germany
| | - Bastian Resch
- Division of Rheumatology, Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Lars-Oliver Tykocinski
- Division of Rheumatology, Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Babak Moradi
- Department of Orthopaedics, Trauma Surgery and Paraplegiology, University of Heidelberg, Heidelberg, Germany
| | - Patrick Horn
- Division of Hematology and Oncology, Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Ziya Kaya
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Norbert Blank
- Division of Rheumatology, Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Stefan Rehart
- Department of Orthopaedics and Trauma Surgery, St Markus Hospital Frankfurt, Frankfurt, Germany
| | - Marc Thomsen
- DRK Clinic for Orthopaedics, Baden-Baden, Germany
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Elena Neumann
- Department of Internal Medicine and Rheumatology, University of Giessen, Kerckhoff-Klinik, Bad Nauheim, Germany
| | - Theresa Tretter
- Division of Rheumatology, Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
16
|
Rituximab Downregulates Gene Expression Associated with Cell Proliferation, Survival, and Proteolysis in the Peripheral Blood from Rheumatoid Arthritis Patients: A Link between High Baseline Autophagy-Related ULK1 Expression and Improved Pain Control. ARTHRITIS 2016; 2016:4963950. [PMID: 27057353 PMCID: PMC4745296 DOI: 10.1155/2016/4963950] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 12/22/2015] [Accepted: 01/03/2016] [Indexed: 12/29/2022]
Abstract
Objective. To clarify molecular mechanisms for the response to rituximab in a longitudinal study. Methods. Peripheral blood from 16 RA patients treated with rituximab for a single treatment course and 26 healthy controls, blood and knee articular cartilages from 18 patients with long-standing RA, and cartilages from 14 healthy subjects were examined. Clinical response was assessed using ESR, ACPA, CRP, RF, DAS28 levels, CD19+ B-cell counts, bone erosion, and joint space narrowing scores. Protein expression in PBMCs was quantified using ELISA. Gene expression was performed with quantitative real-time PCR. Results. A decrease (p < 0.05) in DAS28, ESR, and CRP values after rituximab treatment was associated with the downregulation of MTOR, p21, caspase 3, ULK1, TNFα, IL-1β, and cathepsin K gene expression in the peripheral blood to levels found in healthy subjects. MMP-9 expression remained significantly higher compared to controls although decreased (p < 0.05) versus baseline. A negative correlation between baseline ULK1 gene expression and the number of tender joints at the end of follow-up was observed. Conclusions. The response to rituximab was associated with decreased MTOR, p21, caspase 3, ULK1, TNFα, IL-1β, and cathepsin K gene expression compared to healthy subjects. Residual increased expression in MMP-9, IFNα, and COX2 might account for remaining inflammation and pain. High baseline ULK1 gene expression indicates a good response in respect to pain.
Collapse
|
17
|
Abstract
The immune system is designed to execute rapid, specific, and protective responses against foreign pathogens. To protect against the potentially harmful effects of autoreactive escapees that might arise during the course of the immune response, multiple tolerance checkpoints exist in both the primary and secondary lymphoid organs. Regardless, autoantibodies targeting neural antigens exist in multiple neurologic diseases. The goal of this introductory chapter is to provide a foundation of the major principles and components of the immune system as a framework to understanding autoimmunity and autoimmune neurologic disorders. A broad overview of: (1) innate mechanisms of immunity and their contribution in demyelinating diseases; (2) B and T lymphocytes as effector arms of the adaptive immune response and their contribution to the pathophysiology of neurologic diseases; and (3) emerging therapeutic modalities for treatment of autoimmune disease is provided.
Collapse
Affiliation(s)
- Kay L Medina
- Department of Immunology and Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
18
|
Valor L, López-Longo FJ. [Modulating the survival and maturation system of B lymphocytes: Current and future new therapeutic strategies in systemic lupus erythematosus]. Med Clin (Barc) 2015; 145:206-10. [PMID: 25433780 DOI: 10.1016/j.medcli.2014.08.008] [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: 06/11/2014] [Revised: 07/30/2014] [Accepted: 08/19/2014] [Indexed: 11/25/2022]
Abstract
Systemic lupus erythematosus is an autoimmune disease associated with an aberrant production of autoantibodies by self-reactive B lymphocytes. The study of the phenotypic characteristics of B lymphocytes and the identification of their surface receptors such as BAFF-R, TACI and BCMA, which are responsible of their survival and maturation, have contributed to the development of new therapeutic strategies in recent years.
Collapse
Affiliation(s)
- Lara Valor
- Servicio de Reumatología, Hospital General Universitario Gregorio Marañón, Madrid, España.
| | | |
Collapse
|
19
|
Luo J, Niu X, Zhang M, Zhang K, Chen M, Deng S. Inhibition of B lymphocyte-induced maturation protein-1 reduces the production of autoantibody and alleviates symptoms of systemic lupus erythematosus. Autoimmunity 2015; 48:80-6. [PMID: 25347333 PMCID: PMC4389764 DOI: 10.3109/08916934.2014.976627] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 09/14/2014] [Accepted: 10/05/2014] [Indexed: 12/21/2022]
Abstract
The B lymphocyte-induced maturation protein-1 (Blimp-1) is an important transcription factor for the maintenance of antigen-specific immune responses, and it is crucial in the development of systemic lupus erythematosus (SLE). This study aimed to investigate the role of Blimp-1 in the development of SLE and autoimmune-like symptoms. Lentivirus-mediated Blimp-1 siRNA was constructed and injected into MRL-Fas(lpr) lupus mice. The expression levels of Blimp-1, J-chain, C-myc, XBP-1 and BCMA in peripheral blood mononuclear cells (PMBCs) were determined by RT-PCR. Anti-dsDNA autoantibody levels were detected using ELISA. The expression levels of Blimp-1 in liver, kidney, spleen and lymph nodes of mice were also detected by Western blot. The 24-h urinary protein was monitored weekly. Our results demonstrated that in MRL-Fas(lpr) lupus mice, Blimp-1 was upregulated in PMBCs, liver, kidney, spleen and lymph nodes. Administration of Blimp-1 siRNA reduced the expression of Blimp-1 and the anti-dsDNA level by 78 and 28%, respectively, in the peripheral blood, and the expression of XBP-1, J-chain and BCMA was also decreased. Although the Blimp-1 level in liver showed no significant changes, the levels of Blimp-1 in kidney, spleen and lymph nodes were dramatically decreased by 95, 72 and 47%, respectively. Kidney diseases induced by SLE in lupus mice were mitigated, and urinary protein levels were significantly decreased. These results indicate that Blimp-1 plays an important role in promoting the progression of SLE. Therefore, Blimp-1 may provide a new therapeutic target in the treatment of SLE.
Collapse
MESH Headings
- Animals
- Autoantibodies/biosynthesis
- B-Cell Maturation Antigen/genetics
- B-Cell Maturation Antigen/immunology
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/immunology
- Female
- Gene Expression Regulation
- Genetic Vectors
- Immunoglobulin J-Chains/genetics
- Immunoglobulin J-Chains/immunology
- Injections, Intravenous
- Kidney/immunology
- Kidney/pathology
- Lentivirus/genetics
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/pathology
- Liver/immunology
- Liver/pathology
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/pathology
- Lupus Erythematosus, Systemic/therapy
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Mice
- Mice, Inbred MRL lpr
- Positive Regulatory Domain I-Binding Factor 1
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/immunology
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/genetics
- RNA, Small Interfering/immunology
- Regulatory Factor X Transcription Factors
- Signal Transduction
- Spleen/immunology
- Spleen/pathology
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/genetics
- Transcription Factors/immunology
- X-Box Binding Protein 1
Collapse
Affiliation(s)
- Jie Luo
- Department of Clinical Laboratory, Institute of Surgery Research, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Xiaochang Niu
- Department of Clinical Laboratory, Institute of Surgery Research, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Mingxu Zhang
- Department of Clinical Laboratory, Institute of Surgery Research, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Kejun Zhang
- Department of Clinical Laboratory, Institute of Surgery Research, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Ming Chen
- Department of Clinical Laboratory, Institute of Surgery Research, Daping Hospital, The Third Military Medical University, Chongqing, China
- Address for correspondence: Shaoli Deng, MD and Ming Chen, MD, Department of Clinical Laboratory, Institute of Surgery Research, Daping Hospital, The Third Military Medical University, 10 Chang Jiang Zhi Road, Chongqing 400042, China. E-mail address: (S.D.); (M.C.)
| | - Shaoli Deng
- Department of Clinical Laboratory, Institute of Surgery Research, Daping Hospital, The Third Military Medical University, Chongqing, China
- Address for correspondence: Shaoli Deng, MD and Ming Chen, MD, Department of Clinical Laboratory, Institute of Surgery Research, Daping Hospital, The Third Military Medical University, 10 Chang Jiang Zhi Road, Chongqing 400042, China. E-mail address: (S.D.); (M.C.)
| |
Collapse
|
20
|
Lee J, Sim JH, Kim IJ. Peripheral immature B cells: modulators of autoimmunity. Int J Rheum Dis 2014; 18:200-7. [DOI: 10.1111/1756-185x.12432] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Jisoo Lee
- Division of Rheumatology; Department of Internal Medicine; Ewha Womans University School of Medicine; Seoul South Korea
| | - Ji-Hyun Sim
- Department of Anatomy; Seoul National University College of Medicine; Seoul South Korea
| | - In-Je Kim
- Division of Rheumatology; Department of Internal Medicine; Ewha Womans University School of Medicine; Seoul South Korea
| |
Collapse
|
21
|
Lykken JM, DiLillo DJ, Weimer ET, Roser-Page S, Heise MT, Grayson JM, Weitzmann MN, Tedder TF. Acute and chronic B cell depletion disrupts CD4+ and CD8+ T cell homeostasis and expansion during acute viral infection in mice. THE JOURNAL OF IMMUNOLOGY 2014; 193:746-56. [PMID: 24928986 DOI: 10.4049/jimmunol.1302848] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
B cells provide humoral protection against pathogens and promote cellular immunity through diverse nonclassical effector functions. To assess B cell function in promoting T cell homeostasis, mature B cells were either acutely or chronically depleted in mice using CD20 mAb. Acute B cell depletion in either 2- or 4-mo-old mice significantly reduced spleen and lymph node CD4(+) and CD8(+) T cell numbers, including naive, activated, and Foxp3(+)CD25(+)CD4(+) regulatory T cell subsets. The numbers of IFN-γ- and TNF-α-producing T cells were also significantly reduced. Chronic B cell depletion for 6 mo in aged naive mice resulted in a 40-70% reduction in activated CD4(+) and CD8(+) T cell numbers and 20-50% reductions in IFN-γ-producing T cells. Therefore, B cells were necessary for maintaining naive CD4(+) and CD8(+) T cell homeostasis for subsequent optimal T cell expansion in young and old mice. To determine the significance of this finding, a week of B cell depletion in 4-mo-old mice was followed by acute viral infection with lymphocytic choriomeningitis virus Armstrong. Despite their expansion, activated and cytokine-producing CD4(+) and CD8(+) T cell numbers were still significantly reduced 1 wk later. Moreover, viral peptide-specific CD4(+) and CD8(+) T cell numbers and effector cell development were significantly reduced in mice lacking B cells, whereas lymphocytic choriomeningitis virus titers were dramatically increased. Thus, T cell function is maintained in B cell-depleted mice, but B cells are required for optimal CD4(+) and CD8(+) T cell homeostasis, activation, and effector development in vivo, particularly during responses to acute viral infection.
Collapse
Affiliation(s)
- Jacquelyn M Lykken
- Department of Immunology, Duke University Medical Center, Durham, NC 27710
| | - David J DiLillo
- Department of Immunology, Duke University Medical Center, Durham, NC 27710
| | - Eric T Weimer
- Department of Immunology, Duke University Medical Center, Durham, NC 27710
| | - Susanne Roser-Page
- Atlanta Department of Veterans Affairs Medical Center, Decatur, GA 30033
| | - Mark T Heise
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Jason M Grayson
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157; and
| | - M Neale Weitzmann
- Atlanta Department of Veterans Affairs Medical Center, Decatur, GA 30033; Division of Endocrinology, Metabolism and Lipids, Emory University School of Medicine, Atlanta, GA 30322
| | - Thomas F Tedder
- Department of Immunology, Duke University Medical Center, Durham, NC 27710;
| |
Collapse
|
22
|
Breton CS, Nahimana A, Aubry D, Macoin J, Moretti P, Bertschinger M, Hou S, Duchosal MA, Back J. A novel anti-CD19 monoclonal antibody (GBR 401) with high killing activity against B cell malignancies. J Hematol Oncol 2014; 7:33. [PMID: 24731302 PMCID: PMC4021825 DOI: 10.1186/1756-8722-7-33] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 04/07/2014] [Indexed: 01/16/2023] Open
Abstract
Background CD19 is a B cell lineage specific surface receptor whose broad expression, from pro-B cells to early plasma cells, makes it an attractive target for the immunotherapy of B cell malignancies. In this study we present the generation of a novel humanized anti-CD19 monoclonal antibody (mAb), GBR 401, and investigate its therapeutic potential on human B cell malignancies. Methods GBR 401 was partially defucosylated in order to enhance its cytotoxic function. We analyzed the in vitro depleting effects of GBR 401 against B cell lines and primary malignant B cells from patients in the presence or in absence of purified NK cells isolated from healthy donors. In vivo, the antibody dependent cellular cytotoxicity (ADCC) efficacy of GBR 401 was assessed in a B cell depletion model consisting of SCID mice injected with healthy human donor PBMC, and a malignant B cell depletion model where SCID mice are xenografted with both primary human B-CLL tumors and heterologous human NK cells. Furthermore, the anti-tumor activity of GBR 401 was also evaluated in a xenochimeric mouse model of human Burkitt lymphoma using mice xenografted intravenously with Raji cells. Pharmacological inhibition tests were used to characterize the mechanism of the cell death induced by GBR 401. Results GBR 401 exerts a potent in vitro and in vivo cytotoxic activity against primary samples from patients representing various B-cell malignancies. GBR 401 elicits a markedly higher level of ADCC on primary malignant B cells when compared to fucosylated similar mAb and to Rituximab, the current anti-CD20 mAb standard immunotherapeutic treatment for B cell malignancies, showing killing at 500 times lower concentrations. Of interest, GBR 401 also exhibits a potent direct killing effect in different malignant B cell lines that involves homotypic aggregation mediated by actin relocalization. Conclusion These results contribute to consolidate clinical interest in developing GBR 401 for treatment of hematopoietic B cell malignancies, particularly for patients refractory to anti-CD20 mAb therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Michel A Duchosal
- Service and Central Laboratory of Hematology, University Hospital of Lausanne, Rue du Bugnon 46, 1011- CHUV, Lausanne, Switzerland.
| | | |
Collapse
|
23
|
Nishida S, Kawasaki T, Kashiwagi H, Morishima A, Hishitani Y, Kawai M, Hirano T, Ishii T, Hagihara K, Shima Y, Narazaki M, Ogata A, Oka Y, Kishimoto T, Tanaka T. Successful treatment of acquired hemophilia A, complicated by chronic GVHD, with tocilizumab. Mod Rheumatol 2014. [DOI: 10.3109/s10165-010-0411-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
24
|
Use of rituximab in histologically confirmed idiopathic inflammatory myositis: a case series. Clin Rheumatol 2013; 34:371-7. [DOI: 10.1007/s10067-013-2449-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 11/24/2013] [Indexed: 10/25/2022]
|
25
|
Xu X, Shi Y, Cai Y, Zhang Q, Yang F, Chen H, Gu Y, Zhang M, Yu L, Yang T. Inhibition of increased circulating Tfh cell by anti-CD20 monoclonal antibody in patients with type 1 diabetes. PLoS One 2013; 8:e79858. [PMID: 24278195 PMCID: PMC3835920 DOI: 10.1371/journal.pone.0079858] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 09/26/2013] [Indexed: 01/08/2023] Open
Abstract
Objectives Follicular helper T (Tfh) cells exert an important role in autoimmune diseases. Whether it might be involved in type 1 diabetes (T1D) is unknown. Our aim was to investigate the role of Tfh cells in patients with T1D and the effect of anti-CD20 monoclonal antibody (rituximab) on Tfh cells from T1D patients. Patients and Methods Fifty-four patients with T1D and 37 healthy controls were enrolled in the current study. 20 of those patients were treated with rituximab. The frequencies of circulating CD4+CXCR5+ICOS+T cells were analyzed by flow cytometry. The serum autoantibodies were detected by radioligand assay. The levels of IL-21, IL-6 and BCL-6 were assessed using ELISA and/or real-time PCR. Results Increased frequencies of circulating Tfh cells together with enhanced expression of IL-21 were detected in patients. The correlation between the frequencies of circulating Tfh cells and the serum autoantibodies or C-peptide level was comfirmed. After rituximab therapy, follow-up analysis demonstrated that the frequencies of circulating Tfh cell and serum IA2A were decreased. The levels of IL-21, IL-6 and Bcl-6 mRNA were decreased after treatment. Furthermore, beta cell function in 10 of 20 patients was improved. Conclusions These data indicate Tfh cells may participate in the T1D-relatede immune responses and B cells might play a role in the development of Tfh responses in the disease progression.
Collapse
Affiliation(s)
- Xinyu Xu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yun Shi
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yun Cai
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Qingqing Zhang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Fan Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Heng Chen
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yong Gu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Mei Zhang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Liping. Yu
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- * E-mail:
| |
Collapse
|
26
|
Luu VP, Vazquez MI, Zlotnik A. B cells participate in tolerance and autoimmunity through cytokine production. Autoimmunity 2013; 47:1-12. [DOI: 10.3109/08916934.2013.856006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
27
|
Mollo SB, Zajac AJ, Harrington LE. Temporal requirements for B cells in the establishment of CD4 T cell memory. THE JOURNAL OF IMMUNOLOGY 2013; 191:6052-9. [PMID: 24218454 DOI: 10.4049/jimmunol.1302033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
CD4 T cell memory generation is shaped by a number of factors, including the strength and duration of TCR signaling, as well as the priming environment, all of which can be modified by B cells. Studies using B cell-deficient mice indicate B cells play a critical role in generating effector and memory CD4 T cells; however, when and how B cells are acting to promote these responses has not yet been ascertained. In this study, we use anti-CD20 Ab depletion of B cells at different times following Listeria monocytogenes infection to show that B cells are necessary for the induction of optimal CD4 T cell memory, but not for the transition and maintenance of this population. Importantly, the prerequisite of B cells early postinfection is partially dependent on their expression of MHC class II. B cells are not only required during the priming phase, but also necessary for the initiation of robust secondary responses by memory CD4 T cells. Interestingly, the requirement during the recall response is independent of B cell Ag presentation. Overall, these studies demonstrate the temporally and functionally distinct roles for B cells in regulating CD4 T cell responses.
Collapse
Affiliation(s)
- Sarah B Mollo
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | | | | |
Collapse
|
28
|
Mantani PT, Ljungcrantz I, Andersson L, Alm R, Hedblad B, Björkbacka H, Nilsson J, Fredrikson GN. Circulating CD40+ and CD86+ B cell subsets demonstrate opposing associations with risk of stroke. Arterioscler Thromb Vasc Biol 2013; 34:211-8. [PMID: 24202305 DOI: 10.1161/atvbaha.113.302667] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Accumulating evidence shows that immune cells play an important role in atherosclerosis. Most attention has focused on the role of different T cell subsets, whereas the possible involvement of B cells has been less studied. In this study, we assessed the association of 2 different B cell subsets, CD19(+)CD40(+) and CD19(+)CD86(+) B cells, with risk for development of acute cardiovascular events. APPROACH AND RESULTS The prospective study included 700 subjects randomly selected from the cardiovascular cohort of the Malmö Diet and Cancer study. Mononuclear leukocytes, stored at -140(○)C at the baseline investigation in 1991-1994, were thawed and B cell subsets analyzed by flow cytometry. Cytokine release from CD3/CD28-stimulated mononuclear leukocytes was measured with multiplex ELISA. Baseline carotid intima-media thickness and stenosis were assessed by ultrasonography, and clinical events were monitored through validated national registers during a median/mean follow-up time of 15 years. The subjects in the highest tertile of CD19(+)CD40(+) B cells had a significantly lower risk of incident stroke after adjustment for other risk factors. In contrast, CD19(+)CD86(+) B cells were associated with higher risk for development of a stroke event and increased release of proinflammatory cytokines from mononuclear leukocytes. CONCLUSIONS These observations provide evidence for an involvement of B cells in the incidence of stroke and suggest that both pathogenic and protective B cell subsets exist.
Collapse
Affiliation(s)
- Polyxeni T Mantani
- From the Department of Clinical Sciences, Skåne University Hospital Malmö, Lund University, Malmö, Sweden (P.T.M., I.L., L.A., R.A., B.H., H.B., J.N., G.N.F.); and Faculty of Health and Society, Malmö University, Malmö, Sweden (G.N.F.)
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Feldman AG, Tucker RM, Fenner EK, Pelanda R, Mack CL. B cell deficient mice are protected from biliary obstruction in the rotavirus-induced mouse model of biliary atresia. PLoS One 2013; 8:e73644. [PMID: 23991203 PMCID: PMC3749125 DOI: 10.1371/journal.pone.0073644] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 07/19/2013] [Indexed: 02/04/2023] Open
Abstract
A leading theory regarding the pathogenesis of biliary atresia (BA) is that bile duct injury is initiated by a virus infection, followed by an autoimmune response targeting bile ducts. In experimental models of autoimmune diseases, B cells have been shown to play an important role. The aim of this study was to determine the role of B cells in the development of biliary obstruction in the Rhesus rotavirus (RRV)-induced mouse model of BA. Wild-type (WT) and B cell-deficient (Ig-α(-/-)) mice received RRV shortly after birth. Ig-α(-/-) RRV-infected mice had significantly increased disease-free survival rate compared to WT RRV-infected BA mice (76.8% vs. 17.5%). In stark contrast to the RRV-infected BA mice, the RRV-infected Ig-α(-/-) mice did not have hyperbilirubinemia or bile duct obstruction. The RRV-infected Ig-α(-/-) mice had significantly less liver inflammation and Th1 cytokine production compared to RRV-infected WT mice. In addition, Ig-α(-/-) mice had significantly increased numbers of regulatory T cells (Tregs) at baseline and after RRV infection compared to WT mice. However, depletion of Tregs in Ig-α(-/-) mice did not induce biliary obstruction, indicating that the expanded Tregs in the Ig-α(-/-) mice were not the sole reason for protection from disease. Conclusion : B cell deficient Ig-α(-/-) mice are protected from biliary obstruction in the RRV-induced mouse model of BA, indicating a primary role of B cells in mediating disease pathology. The mechanism of protection may involve lack of B cell antigen presentation, which impairs T-cell activation and Th1 inflammation. Immune modulators that inhibit B cell function may be a new strategy for treatment of BA.
Collapse
Affiliation(s)
- Amy G Feldman
- Department of Pediatrics, Section of Pediatric Gastroenterology, Children's Hospital, Colorado, Aurora, Colorado, USA.
| | | | | | | | | |
Collapse
|
30
|
Stalika E, Kanellis G, Papalexandri A, Iskas M, Vrakidou E, Demonakou M, Anagnostopoulos A, Stamatopoulos K, Papadaki T. Cytotoxic T cell-mediated gastritis after rituximab treatment for gastric malt lymphoma. Leuk Lymphoma 2013; 55:702-5. [DOI: 10.3109/10428194.2013.810736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
31
|
Finch DK, Ettinger R, Karnell JL, Herbst R, Sleeman MA. Effects of CXCL13 inhibition on lymphoid follicles in models of autoimmune disease. Eur J Clin Invest 2013; 43:501-9. [PMID: 23517338 DOI: 10.1111/eci.12063] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 02/05/2013] [Indexed: 12/22/2022]
Abstract
The chemokine CXCL13 has a key role in secondary lymphoid tissue orchestration and lymphoid neogenesis. Transgenic mice deficient in CXCL13 or its receptor CXCR5 have severely impaired lymph node development, lack peritoneal B-lymphocytes and are deficient in circulating antibodies to common bacterial antigens. However, total circulating numbers of B-lymphocytes are slightly elevated and humoral responses to T-dependent or blood-borne antigens are relatively normal. Lymphoid neogenesis is an aberrant process that occurs in chronically inflamed tissue and provides a microenvironment supportive of pathogenic B-cell survival and activation. Here, we describe the impact of therapeutic dosing of a CXCL13 antibody in a mouse model of arthritis, and detail the contribution CXCL13 makes to lymphoid follicle microenvironment, without affecting humoral immune responses.
Collapse
|
32
|
Blüml S, McKeever K, Ettinger R, Smolen J, Herbst R. B-cell targeted therapeutics in clinical development. Arthritis Res Ther 2013; 15 Suppl 1:S4. [PMID: 23566679 PMCID: PMC3624127 DOI: 10.1186/ar3906] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
B lymphocytes are the source of humoral immunity and are thus a critical component of the adaptive immune system. However, B cells can also be pathogenic and the origin of disease. Deregulated B-cell function has been implicated in several autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, and multiple sclerosis. B cells contribute to pathological immune responses through the secretion of cytokines, costimulation of T cells, antigen presentation, and the production of autoantibodies. DNA-and RNA-containing immune complexes can also induce the production of type I interferons, which further promotes the inflammatory response. B-cell depletion with the CD20 antibody rituximab has provided clinical proof of concept that targeting B cells and the humoral response can result in significant benefit to patients. Consequently, the interest in B-cell targeted therapies has greatly increased in recent years and a number of new biologics exploiting various mechanisms are now in clinical development. This review provides an overview on current developments in the area of B-cell targeted therapies by describing molecules and subpopulations that currently offer themselves as therapeutic targets, the different strategies to target B cells currently under investigation as well as an update on the status of novel therapeutics in clinical development. Emerging data from clinical trials are providing critical insight regarding the role of B cells and autoantibodies in various autoimmune conditions and will guide the development of more efficacious therapeutics and better patient selection.
Collapse
Affiliation(s)
- Stephan Blüml
- MedImmune, LLC, Department of Research, One MedImmune Way, Gaithersburg, MD 20854, USA
| | | | | | | | | |
Collapse
|
33
|
Váncsa A, Szabó Z, Szamosi S, Bodnár N, Végh E, Gergely L, Szucs G, Szántó S, Szekanecz Z. Longterm effects of rituximab on B cell counts and autoantibody production in rheumatoid arthritis: use of high-sensitivity flow cytometry for more sensitive assessment of B cell depletion. J Rheumatol 2013; 40:565-71. [PMID: 23547216 DOI: 10.3899/jrheum.111488] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To assess the efficacy and safety of longterm rituximab (RTX) therapy for rheumatoid arthritis (RA) and study correlations among B cell depletion, clinical response, and autoantibody production. METHODS Seventy-seven patients with moderate or high RA activity received RTX and were re-treated every 6 months regardless of clinical response. All patients received at least 5 cycles. We assessed 28-joint Disease Activity Score (DAS28), IgM rheumatoid factor (RF), and anticitrullinated protein antibody (ACPA) levels at baseline, after 15 days, and then every 6 months for 24 months. Absolute CD19+ B lymphocyte counts were determined in 50 patients using high-sensitivity flow cytometry (hsFACS) by reading 100,000 events. RESULTS After 6, 12, 18, and 24 months, 51.6%, 51.9%, 73.3%, and 83.8% of patients, respectively, showed good European League Against Rheumatism responses. Significant and sustained decreases in IgM RF and ACPA levels were observed as early as 6 months and 12 months, respectively. The baseline mean absolute B cell number was 0.234 g/l. B cell numbers diminished significantly after the very first infusion by Day 15 (0.104 g/l; p = 0.007); they further decreased until 24 months (0.0013 g/l; p < 0.001). One RTX infusion resulted in incomplete depletion in 76.7% of patients. Upon RTX treatment, changes in CD19+ B cell numbers positively correlated with changes in DAS28 (r = 0.963, p = 0.008) and IgM RF (r = 0.859, p = 0.028), but not with changes in ACPA production (r = 0.726, p = 0.102). The correlations between B cell numbers and DAS28 were observed in both ACPA-seropositive (r = 0.999, p < 0.0001) and ACPA-negative patient subpopulations (r = 0.962, p = 0.009). The correlation between CD19+ cell numbers and IgM RF was observed only in the ACPA-positive population (r = 0.944, p = 0.005) but not in seronegative patients (r = 0.398, p = 0.435). No safety issues arose. CONCLUSION In RA, clinical response to RTX is associated with the extent of B cell depletion and with autoantibody production. Changes in CD19+ B cell numbers correlate with those in disease activity and, in seropositive patients, also with IgM RF, but not with ACPA production. We found that hsFACS may be a useful method to more accurately assess incomplete B cell depletion.
Collapse
Affiliation(s)
- Andrea Váncsa
- Department of Rheumatology, Division of Hematology, Institute of Medicine, University of Debrecen Medical and Health Sciences Center, Debrecen, Hungary.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Blimp-1 siRNA inhibits B cell differentiation and prevents the development of lupus in mice. Hum Immunol 2012; 74:297-301. [PMID: 23220434 DOI: 10.1016/j.humimm.2012.11.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 11/14/2012] [Accepted: 11/27/2012] [Indexed: 01/05/2023]
Abstract
Cumulative evidence suggest that B-lymphocytes play a role in the development of systemic lupus erythematosus (SLE). Thus, the therapeutic approach targeting specific B cells provides a promising way to treat SLE. Blimp-1 (B lymphocyte induced maturation protein), a transcriptional factor, controls the terminal differentiation of mature B cells to plasma cells. To explore the potential of Blimp-1 in the SLE development, we constructed the adenovirus encoding Blimp-1 siRNA, and injected it into BWF1 lupus mice. The results demonstrated that Blimp-1 siRNA decreased the Blimp-1 expression of B cells by regulating XBP-1 (X Box binding protein-1), BCMA (B-cell maturation antigen) expression through c-myc pathway. In addition, Blimp-1 siRNA eliminated anti-dsDNA antibody-producing plsma cells, reduced serum anti-dsDNA antibody levels and impeded the development of lupus. Therefore, our data provide the insight into the mechanism of Blimp-1 in SLE development and might represent a promising therapeutic strategy for autoantibody-mediated diseases.
Collapse
|
35
|
Cianchini G, Lupi F, Masini C, Corona R, Puddu P, De Pità O. Therapy with rituximab for autoimmune pemphigus: Results from a single-center observational study on 42 cases with long-term follow-up. J Am Acad Dermatol 2012; 67:617-22. [DOI: 10.1016/j.jaad.2011.11.007] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 10/29/2011] [Accepted: 11/06/2011] [Indexed: 11/15/2022]
|
36
|
Affiliation(s)
- Sarah Lawrie
- Neuroimmunology Unit; Montreal Neurological Institute; McGill University; Montreal; QC; Canada
| | | |
Collapse
|
37
|
León B, Ballesteros-Tato A, Misra RS, Wojciechowski W, Lund FE. Unraveling effector functions of B cells during infection: the hidden world beyond antibody production. Infect Disord Drug Targets 2012; 12:213-21. [PMID: 22394173 PMCID: PMC4517595 DOI: 10.2174/187152612800564437] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 02/25/2012] [Indexed: 12/12/2022]
Abstract
Antibodies made by B cells are critically important for immune protection to a variety of infectious agents. However, it is becoming increasingly clear that B cells do more than make antibodies and that B cells can both enhance and suppress immune responses. Furthermore, there is growing evidence that B cells modulate cellular immune responses by antibody dependent and independent mechanisms. Although we have a good understanding of the roles played by antibody- secreting effector B cells during immune responses, we know very little about the Ab independent "effector" functions of B cells in either health or disease. Given the recent data suggesting that B cells may contribute to autoimmune disease pathogenesis via an antibody independent mechanism and the increasing use of B cell depletion therapy in autoimmune patients, investigators are beginning to reassess the multiple roles for B cells during immune responses. In this article, we review data describing how B cells mediate protection to pathogens independently of antibody production. In particular, we will focus on the role that B cells play in facilitating dendritic cell and T cell interactions in lymph nodes, the importance of antigen-presenting B cells in sustaining effector T cell and T follicular helper responses to pathogens and the relevance of cytokine-producing effector and regulatory B cells in immune responses.
Collapse
Affiliation(s)
- Beatriz León
- University of Alabama, Dept. of Microbiology, Birmingham, AL 35294-2170, USA
| | | | | | | | | |
Collapse
|
38
|
|
39
|
Iseki M, Omori-Miyake M, Xu W, Sun X, Takaki S, Rawlings DJ, Ziegler SF. Thymic stromal lymphopoietin (TSLP)-induced polyclonal B-cell activation and autoimmunity are mediated by CD4+ T cells and IL-4. Int Immunol 2012; 24:183-95. [PMID: 22281511 DOI: 10.1093/intimm/dxr113] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The cytokine thymic stromal lymphopoietin (TSLP) functions as a regulator of bone marrow B-cell development and a key initiator of allergic inflammation. In the current study, we show that mature B cells, derived from transgenic mice with systemically elevated levels of TSLP (K5-TSLP mice), exhibit markedly enhanced mitogenic responses in vitro and that this enhanced responsiveness leads to polyclonal B-cell activation and development of autoimmune hemolytic anemia in vivo. In contrast, B cells derived from K5-TSLP mice lacking CD4(+) T cells failed to show polyclonal activation. Furthermore, neither mature B-cell activation nor hemolytic anemia occurred in IL-4-deficient K5-TSLP mice. Consistent with these findings, activation of mature B cells occurred independently of B-cell intrinsic TSLP signals. Taken together, our results demonstrate that systemic alterations in TSLP, through induction of IL-4 from CD4(+) T cells and other cell types, functions as an important factor in peripheral B-cell homeostasis and promotion of humoral autoimmunity.
Collapse
|
40
|
Roccatello D, Sciascia S, Rossi D, Alpa M, Naretto C, Baldovino S, Menegatti E, La Grotta R, Modena V. Intensive short-term treatment with rituximab, cyclophosphamide and methylprednisolone pulses induces remission in severe cases of SLE with nephritis and avoids further immunosuppressive maintenance therapy. Nephrol Dial Transplant 2011; 26:3987-3992. [DOI: 10.1093/ndt/gfr109] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
41
|
Feldman RJ, Ahmed AR. Relevance of rituximab therapy in pemphigus vulgaris: analysis of current data and the immunologic basis for its observed responses. Expert Rev Clin Immunol 2011; 7:529-41. [PMID: 21790294 DOI: 10.1586/eci.11.22] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Treatment of pemphigus vulgaris (PV) patients with rituximab therapy has not been critically evaluated. This article will provide in significant detail the available data to date, in order to provide a clinical and immunologic basis for clinicians to decide how best to treat recalcitrant PV patients with rituximab. PV is an autoimmune blistering disorder that affects the skin and mucous membranes. The immunopathology is well characterized, including the target antigens. PV patients have traditionally been treated with systemic corticosteroids and adjuvant immunosuppressive therapies. Clinical remission has been achieved in roughly 30% of patients. However, many patients experience severe side effects from this immunosuppression, including death. B-cell depletion therapy with rituximab therapy has been used to treat several autoimmune diseases including PV. In this article, we examined the data on 153 patients with PV who have been treated with rituximab. Our focus is on the clinical response of the patients with emphasis on adjuvant therapies, dosing regimens, potential adverse events and mechanism of action related to B-cell modulation during therapy. Importantly, the use of rituximab has increased clinical remission rates to 65% including many patients who were able to discontinue all systemic medications. Finally, an expert commentary is provided, which includes suggestions for optimizing current therapy and recommends the future direction of the field. The authors strongly endorse the use of rituximab in treatment of PV patients, particularly those nonresponsive to or who develop serious side effects to conventional therapy. Proper monitoring of patients including peripheral B-cell counts and overt signs of infection are warranted, given the potential for prolonged B-cell depletion.
Collapse
Affiliation(s)
- Ron J Feldman
- Center for Blistering Diseases, 70 Parker Hill Avenue, Boston, MA 02120, USA
| | | |
Collapse
|
42
|
Abstract
Prevention of loss of b cells in type 1 diabetes is a major goal of current research. Knowledge of the genetic susceptibility, increasing ability to predict who may be at risk, recognition of the potential clinical impact of residual insulin secretion after diagnosis, and development of new immunomodulatory agents have supported an increasing number of clinical trials to prevent b-cell loss. Interventions can be targeted at 3 stages: before the development of autoimmunity (primary prevention), after autoimmunity is recognized (secondary prevention), or after diagnosis when significant numbers of b cells remain (tertiary prevention). Thus far, several agents show promise when given shortly after diagnosis, but no interventions before diagnosis have shown benefit. Knowledge in this area has grown quickly in recent years and will continue to grow rapidly with several international collaborative efforts underway.
Collapse
Affiliation(s)
- Diane K Wherrett
- Division of Endocrinology, Department of Pediatrics, The Hospital for Sick Children and University of Toronto, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada
| | | |
Collapse
|
43
|
Abstract
B-lymphocytes have traditionally been thought to contribute to immunity and autoimmune disease through terminal differentiation into plasma cells that secrete antibody. However, studies in mice and recent clinical studies have demonstrated that genetically altered B-cell function and B-cell-targeted therapies can significantly affect autoimmune diseases that were predominantly thought to be T-cell-mediated. B-cell depletion in mouse models of disease has also led to the identification of alternative B-cell effector functions that regulate normal immune responses and autoimmune disease. This review highlights multiple B-cell effector mechanisms, including the promotion of cellular immunity, the negative regulation of immune responses, and the production of pathogenic antibodies.
Collapse
Affiliation(s)
- David J DiLillo
- Department of Immunology, Duke University Medical Center, Room 353 Jones Building, Research Drive, Box 3010, Durham, NC 27710, USA
| | | | | |
Collapse
|
44
|
Chamberlain JL, Attridge K, Wang CJ, Ryan GA, Walker LSK. B cell depletion in autoimmune diabetes: insights from murine models. Expert Opin Ther Targets 2011; 15:703-14. [PMID: 21366498 PMCID: PMC3997824 DOI: 10.1517/14728222.2011.561320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION The incidence of type 1 diabetes (T1D) is rising for reasons that largely elude us. New strategies aimed at halting the disease process are needed. One type of immune cell thought to contribute to T1D is the B lymphocyte. The first Phase II trial of B cell depletion in new onset T1D patients indicated that this slowed the destruction of insulin-producing pancreatic beta cells. The mechanistic basis of the beneficial effects remains unclear. AREAS COVERED Studies of B cell depletion and deficiency in animal models of T1D. How B cells can influence T cell-dependent autoimmune diabetes in animal models. The heterogeneity of B cell populations and current evidence for the potential contribution of specific B cell subsets to diabetes, with emphasis on marginal zone B cells and B1 B cells. EXPERT OPINION B cells can influence the T cell response to islet antigens and B cell depletion or genetic deficiency is associated with decreased insulitis in animal models. New evidence suggests that B1 cells may contribute to diabetes pathogenesis. A better understanding of the roles of individual B cell subsets in disease will permit fine-tuning of therapeutic strategies to modify these populations.
Collapse
Affiliation(s)
- Jayne L Chamberlain
- University of Birmingham Medical School, School of Immunity & Infection, IBR Building, Birmingham B15 2TT, UK
| | - Kesley Attridge
- University of Birmingham Medical School, School of Immunity & Infection, IBR Building, Birmingham B15 2TT, UK
| | - Chun Jing Wang
- University of Birmingham Medical School, School of Immunity & Infection, IBR Building, Birmingham B15 2TT, UK
| | - Gemma A Ryan
- University of Birmingham Medical School, School of Immunity & Infection, IBR Building, Birmingham B15 2TT, UK
| | - Lucy SK Walker
- University of Birmingham Medical School, Medical Research Council Center for Immune Regulation, Birmingham B15 2TT, UK
| |
Collapse
|
45
|
Targeting siglecs--a novel pharmacological strategy for immuno- and glycotherapy. Biochem Pharmacol 2011; 82:323-32. [PMID: 21658374 DOI: 10.1016/j.bcp.2011.05.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 05/17/2011] [Indexed: 01/13/2023]
Abstract
The immune system must be tightly held in check to avoid bystander tissue damage as well as autoreactivity caused by overwhelming immune reactions. A novel family of immunoregulatory, carbohydrate-binding receptors, the Siglecs (sialic acid binding immunoglobulin-like lectins), has received particular attention in light of their capacity to mediate cell death, anti-proliferative effects and to regulate a variety of cellular activities. Siglec receptors are mainly expressed on leukocytes in a cell type-specific and differentiation-dependent manner. Siglecs might potentially be exploited as targets of novel immune- and glycotherapeutics for cell-directed therapies in autoimmune and allergic diseases, as well as in haematologic malignancies. Here we present novel insights on structural and functional characteristics, expression patterns and evolutionary aspects of Siglecs and their ligands. Pharmacological strategies using Siglec agonistic cross-linking therapeutics, such as monoclonal or engineered antibodies, intravenous immunoglobulin (IVIG), or glycomimetics are discussed. Modulation of immune responses by targeting Siglecs using agonistic or antagonistic therapeutics may have important clinical implications and may pave the way for novel pharmacological avenues for the treatment of autoimmune and allergic diseases or for tumor immunotherapy.
Collapse
|
46
|
Neves M, Alves JD. Factors implicated in the generation and persistence of long-lived plasma cell-mediated autoimmunity. Autoimmun Rev 2011; 10:375-82. [DOI: 10.1016/j.autrev.2010.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Accepted: 12/20/2010] [Indexed: 12/21/2022]
|
47
|
Nishida S, Kawasaki T, Kashiwagi H, Morishima A, Hishitani Y, Kawai M, Hirano T, Ishii T, Hagihara K, Shima Y, Narazaki M, Ogata A, Oka Y, Kishimoto T, Tanaka T. Successful treatment of acquired hemophilia A, complicated by chronic GVHD, with tocilizumab. Mod Rheumatol 2011; 21:420-2. [PMID: 21240617 DOI: 10.1007/s10165-010-0411-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 12/18/2010] [Indexed: 11/30/2022]
Abstract
A 65-year-old woman who had suffered from chronic graft-versus-host disease (GVHD) presented with extensive purpura and was diagnosed with acquired hemophilia A. Because she was refractory to corticosteroids and her condition was complicated with diabetes mellitus, glaucoma, and hypoglobulinemia, she was treated with tocilizumab. Tocilizumab treatment increased the activity of factor VIII in a rapid and sustained manner, leading to a reduction of the prednisolone dose. Tocilizumab may thus be an optional treatment modality for acquired hemophilia A.
Collapse
Affiliation(s)
- Sumiyuki Nishida
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Deng S, Yuan T, Cheng X, Jian R, Jiang J. B-lymphocyte-induced maturation protein1 up-regulates the expression of B-cell maturation antigen in mouse plasma cells. Mol Biol Rep 2010; 37:3747-3755. [PMID: 20339926 DOI: 10.1007/s11033-010-0028-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2009] [Accepted: 02/24/2010] [Indexed: 11/26/2022]
Abstract
B-lymphocyte-induced maturation protein1(Blimp-1) and B-cell maturation antigen (BCMA) are essential factors in the development and survival of plasma cells. However, whether Blimp-1 could regulate the expression of BCMA is unknown. We found that the BCMA promoter region did not have typical "TATA" and "CAAT" box, but contained several potential binding sites of transcription factors, including the consensus sequences for Blimp-1, located in the "-31 to -21" and "-46 to -36" from the potential transcription initiation site of the mouse BCMA gene, respectively. Furthermore, induction of Blimp-1 over-expression significantly up-regulated the expression of BCMA and increased the BCMA promoter activity in mouse J558L plasma cells. In parallel, knockdown of Blimp-1 expression by the Blimp-1-specific shRNA significantly reduced the BCMA mRNA transcription and protein expression in J558L cells in vitro. Substitution mutation of the "-38 to -42" sequence, but not the "-23 to -27", in the BCMA promoter abolished the regulatory effect of Blimp-1 on the expression of BCMA. Importantly, Blimp-1 bound to the "GAAAC", but not its mutant "GATTC", contained BCMA promoter, as determined by competitive electrophoretic mobility shift assay (EMSA). Therefore, our data clearly suggest that Blimp-s a positive regulator of the expression of BCMA gene in mouse plasma cells.
Collapse
Affiliation(s)
- Shaoli Deng
- Department of Clinical Laboratory, Third Affiliated Hospital of Third Military Medical University, Chongqing, 400042, China.
| | | | | | | | | |
Collapse
|
49
|
Characterization of a rare IL-10-competent B-cell subset in humans that parallels mouse regulatory B10 cells. Blood 2010; 117:530-41. [PMID: 20962324 DOI: 10.1182/blood-2010-07-294249] [Citation(s) in RCA: 875] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Regulatory B cells control inflammation and autoimmunity in mice, including the recently identified IL-10-competent B10 cell subset that represents 1% to 3% of spleen B cells. In this study, a comparable IL-10-competent B10 cell subset was characterized in human blood. B10 cells were functionally identified by their ability to express cytoplasmic IL-10 after 5 hours of ex vivo stimulation, whereas progenitor B10 (B10pro) cells required 48 hours of in vitro stimulation before they acquired the ability to express IL-10. B10 and B10pro cells represented 0.6% and approximately 5% of blood B cells, respectively. Ex vivo B10 and B10pro cells were predominantly found within the CD24(hi)CD27(+) B-cell subpopulation that was able to negatively regulate monocyte cytokine production through IL-10-dependent pathways during in vitro functional assays. Blood B10 cells were present in 91 patients with rheumatoid arthritis, systemic lupus erythematosus, primary Sjögren syndrome, autoimmune vesiculobullous skin disease, or multiple sclerosis, and were expanded in some cases as occurs in mice with autoimmune disease. Mean B10 + B10pro-cell frequencies were also significantly higher in patients with autoimmune disease compared with healthy controls. The characterization of human B10 cells will facilitate their identification and the study of their regulatory activities during human disease.
Collapse
|
50
|
Jordan SC, Reinsmoen N, Peng A, Lai CH, Cao K, Villicana R, Toyoda M, Kahwaji J, Vo AA. Advances in diagnosing and managing antibody-mediated rejection. Pediatr Nephrol 2010; 25:2035-45; quiz 2045-8. [PMID: 20077121 PMCID: PMC2923704 DOI: 10.1007/s00467-009-1386-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 10/05/2009] [Accepted: 11/02/2009] [Indexed: 02/07/2023]
Abstract
Antibody-mediated rejection (AMR) is a unique, significant, and often severe form of allograft rejection that is not amenable to treatment with standard immunosuppressive medications. Significant advances have occurred in our ability to predict patients at risk for, and to diagnose, AMR. These advances include the development of newer anti-human leukocyte antigen (HLA)-antibody detection techniques and assays for non-HLA antibodies associated with AMR. The pathophysiology of AMR suggests a prime role for antibodies, B cells and plasma cells, but other effector molecules, especially the complement system, point to potential targets that could modify the AMR process. An emerging and potentially larger problem is the development of chronic AMR (CAMR) resulting from de novo donor-specific anti-HLA antibodies (DSA) that emerge more than 100 days posttransplantation. Therapeutic options include: (1) High-dose intravenously administered immunoglobulin (IVIG), which has many potential benefits. (2) The use of IVIG+rituximab (anti-CD20, anti-B cell). (3) The combination of plasmapheresis (PP)+low-dose IVIG with or without rituximab. Data support the efficacy of all of the above approaches. Newer approaches to treating AMR include using the proteosome inhibitor (bortezomib), which induces apoptosis in plasma cells, and eculizumab (anti-C5, anticomplement monoclonal antibody).
Collapse
Affiliation(s)
- Stanley C Jordan
- The Transplant Immunotherapy Program, Comprehensive Transplant Center, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|