1
|
Stohl W, Wu Y, Stohl M. Contributions of each of the BAFF receptors to the lymphocyte profiles in C57BL/6 mice. Immunology 2024; 173:689-711. [PMID: 39215598 DOI: 10.1111/imm.13856] [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: 03/27/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
BAFF, a vital B cell survival and differentiation factor, has three receptors: B-cell maturation antigen (BCMA), transmembrane activator and CAML interactor (TACI) and BR3. Although B cells are greatly reduced in B6.Baff-/- (which harbour no BAFF) and B6.Br3-/- mice (which harbour supra-normal levels of BAFF), the distributions of B cell subsets and relationships between Foxp3+ and CD4+ cells in these mice differ. Using a large panel of B6 congenic knockout and/or transgenic mice, we demonstrate that (1) supra-normal levels of BAFF per se do not explain the phenotypic differences between B6.Baff-/- and B6.Br3-/- mice; (2) B cells are expanded in B6.Taci-/- mice, with preferential expansion of follicular (FO) B cells at the expense of CD19+CD21-/loCD23-/lo B cells but without the preferential expansion of Foxp3+ cells observed in B6 mice bearing a Baff transgene; (3) despite no expansion in total B cells, percentages of FO B cells and marginal zone B cells are higher and percentages of CD19+CD21-/loCD23-/lo B cells are lower in young B6.Bcma-/- mice, consistent with the inability of B6.Br3-/-.Taci-/- mice to recapitulate the B cell profile of B6.Baff-/- mice; and (4) percentages of Foxp3+ cells in B6.Br3-/-.Taci-/- mice are intermediate between those in B6.Br3-/- and B6.Taci-/- mice despite the B cell profile of B6.Br3-/-.Taci-/- mice strongly resembling that of B6.Br3-/- mice. Collectively, our findings point to a non-redundant role for each of the BAFF receptors in determining the ultimate lymphocyte profile of the host. This may have clinically relevant ramifications in that the degree that a candidate therapeutic agent blocks engagement of any given individual BAFF receptor may affect its clinical utility.
Collapse
Affiliation(s)
- William Stohl
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Ying Wu
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Malka Stohl
- New York State Psychiatric Institute, New York, New York, USA
| |
Collapse
|
2
|
Zeng L, Yang K, Wu Y, Yu G, Yan Y, Hao M, Song T, Li Y, Chen J, Sun L. Telitacicept: A novel horizon in targeting autoimmunity and rheumatic diseases. J Autoimmun 2024; 148:103291. [PMID: 39146891 DOI: 10.1016/j.jaut.2024.103291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 06/19/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
BLyS and APRIL have the capability to bind to B cells within the body, allowing these cells to evade elimination when they should naturally be removed. While BLyS primarily plays a role in B cell development and maturation, APRIL is linked to B cell activation and the secretion of antibodies. Thus, in theory, inhibiting BLyS or APRIL could diminish the population of aberrant B cells that contribute to SLE and reduce disease activity in patients. Telitacicept functions by binding to and neutralizing the activities of both BLyS and APRIL, thus hindering the maturation and survival of plasma cells and fully developed B cells. The design of telitacicept is distinctive; it is not a monoclonal antibody but a TACI-Fc fusion protein generated through recombinant DNA technology. This fusion involves merging gene segments of the TACI protein, which can target BLyS/APRIL simultaneously, with the Fc gene segment of the human IgG protein. The TACI-Fc fusion protein exhibits the combined characteristics of both proteins. Currently utilized for autoimmune disease treatment, telitacicept is undergoing clinical investigations globally to assess its efficacy in managing various autoimmune conditions. This review consolidates information on the mechanistic actions, dosing regimens, pharmacokinetics, efficacy, and safety profile of telitacicept-a dual-targeted biological agent. It integrates findings from prior experiments and pharmacokinetic analyses in the treatment of RA and SLE, striving to offer a comprehensive overview of telitacicept's research advancements.
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China; Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China.
| | - Yang Wu
- Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ganpeng Yu
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Yexing Yan
- Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Moujia Hao
- Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Tian Song
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuwei Li
- School of Mathematics and Computational Science, Hunan University of Science and Technology, Hunan, China
| | - Junpeng Chen
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, USA; Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China; Tong Jiecheng Studio, Hunan University of Science and Technology, Xiangtan, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China; Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
3
|
Pitsiu M, Yalkinoglu Ö, Farrell C, Girard P, Vazquez‐Mateo C, Papasouliotis O. Population pharmacokinetics of atacicept in systemic lupus erythematosus: An analysis of three clinical trials. CPT Pharmacometrics Syst Pharmacol 2023; 12:1157-1169. [PMID: 37332136 PMCID: PMC10431037 DOI: 10.1002/psp4.12982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 06/20/2023] Open
Abstract
B cell stimulating factor (BLyS) and a proliferation-inducing ligand (APRIL) are targets for novel treatments in patients with systemic lupus erythematosus (SLE). Atacicept is a recombinant, soluble fusion protein that blocks BLyS and APRIL activity. This study characterized the pharmacokinetic (PK) profile of atacicept using a population PK model and identified covariates explaining the PK variability. Total atacicept concentrations from a phase I study in healthy volunteers and two phase II studies in patients with SLE, using subcutaneous administration, were modeled using a quasi-steady-state approximation of the target-mediated drug disposition model with first-order absorption. The model included 3640 serum atacicept concentration records from 37 healthy volunteers and 503 patients with SLE and described total atacicept concentrations of the three trials, providing precise estimates of all parameters. Body weight and baseline BLyS concentration were the only statistically significant covariates, whereas no differences were found between patients and healthy volunteers. Apparent clearance and volume of the central compartment increased with body weight and initial target concentration increased with baseline BLyS. The change on atacicept exposure was moderate, with a difference in area under the curve compared with the median of 20%-32% for body weight, and 7%-18% for BLyS. Therefore, the effects of these covariates on atacicept exposure are not expected to be clinically relevant. The model described the complete total atacicept concentration-time profiles without finding any differences between healthy subjects and patients with SLE and supports the 150 mg once weekly dose for further trials.
Collapse
Affiliation(s)
- Maria Pitsiu
- Quantitative Pharmacology and PharmacometricsICON Clinical Research UK LtdMarlowUK
| | - Özkan Yalkinoglu
- Translational Medicinethe healthcare business of Merck KGaADarmstadtGermany
| | - Colm Farrell
- Quantitative Pharmacology and PharmacometricsICON Clinical Research UK LtdMarlowUK
| | - Pascal Girard
- Merck Institute of Pharmacometrics, Lausanne, Switzerland, an Affiliate of Merck KGaA, Darmstadt, GermanyAres Trading S.A.LausanneSwitzerland
| | | | - Orestis Papasouliotis
- Merck Institute of Pharmacometrics, Lausanne, Switzerland, an Affiliate of Merck KGaA, Darmstadt, GermanyAres Trading S.A.LausanneSwitzerland
| |
Collapse
|
4
|
Athanassiou P, Athanassiou L. Current Treatment Approach, Emerging Therapies and New Horizons in Systemic Lupus Erythematosus. Life (Basel) 2023; 13:1496. [PMID: 37511872 PMCID: PMC10381582 DOI: 10.3390/life13071496] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/18/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Systemic lupus erythematosus (SLE), the prototype of systemic autoimmune diseases is characterized by extreme heterogeneity with a variable clinical course. Renal involvement may be observed and affects the outcome. Hydroxychloroquine should be administered to every lupus patient irrespective of organ involvement. Conventional immunosuppressive therapy includes corticosteroids, methotrexate, cyclophosphamide, mycophenolate mofetil, azathioprine, cyclosporine and tacrolimus. However, despite conventional immunosuppressive treatment, flares occur and broad immunosuppression is accompanied by multiple side effects. Flare occurrence, target organ involvement, side effects of broad immunosuppression and increased knowledge of the pathogenetic mechanisms involved in SLE pathogenesis as well as the availability of biologic agents has led to the application of biologic agents in SLE management. Biologic agents targeting various pathogenetic paths have been applied. B cell targeting agents have been used successfully. Belimumab, a B cell targeting agent, has been approved for the treatment of SLE. Rituximab, an anti-CD20 targeting agent is also used in SLE. Anifrolumab, an interferon I receptor-targeting agent has beneficial effects on SLE. In conclusion, biologic treatment is applied in SLE and should be further evaluated with the aim of a good treatment response and a significant improvement in quality of life.
Collapse
Affiliation(s)
| | - Lambros Athanassiou
- Department of Rheumatology, Asclepeion Hospital, Voula, GR16673 Athens, Greece
| |
Collapse
|
5
|
The BAFF-APRIL System in Cancer. Cancers (Basel) 2023; 15:cancers15061791. [PMID: 36980677 PMCID: PMC10046288 DOI: 10.3390/cancers15061791] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/13/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
B cell-activating factor (BAFF; also known as CD257, TNFSF13B, BLyS) and a proliferation-inducing ligand (APRIL; also known as CD256, TNFSF13) belong to the tumor necrosis factor (TNF) family. BAFF was initially discovered as a B-cell survival factor, whereas APRIL was first identified as a protein highly expressed in various cancers. These discoveries were followed by over two decades of extensive research effort, which identified overlapping signaling cascades between BAFF and APRIL, controlling immune homeostasis in health and driving pathogenesis in autoimmunity and cancer, the latter being the focus of this review. High levels of BAFF, APRIL, and their receptors have been detected in different cancers and found to be associated with disease severity and treatment response. Here, we have summarized the role of the BAFF-APRIL system in immune cell differentiation and immune tolerance and detailed its pathogenic functions in hematological and solid cancers. We also highlight the emerging therapeutics targeting the BAFF-APRIL system in different cancer types.
Collapse
|
6
|
Eslami M, Willen D, Papasouliotis O, Schuepbach-Mallpell S, Willen L, Donzé O, Yalkinoglu Ö, Schneider P. Kinetics of free and ligand-bound atacicept in human serum. Front Immunol 2022; 13:1035556. [PMID: 36532058 PMCID: PMC9756848 DOI: 10.3389/fimmu.2022.1035556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
BAFF (B cell activation factor of the TNF family/B lymphocyte stimulator, BLyS) and APRIL (a proliferation-inducing ligand) are targeted by atacicept, a decoy receptor consisting of the extracellular domain of TACI (transmembrane activator and calcium-modulator and cyclophilin (CAML) interactor) fused to the Fc portion of human IgG1. The purpose of the study was to characterize free and ligand-bound atacicept in humans. Total and active atacicept in serum of healthy volunteers receiving a single dose of subcutaneous atacicept or in patients treated weekly for one year were measured by ELISA, Western blot, or cell-based assays. Pharmacokinetics of free and bound atacicept were predicted based on total atacicept ELISA results. Persistence of complexes of purified atacicept bound to recombinant ligands was also monitored in mice. Results show that unbound or active atacicept in human serum exceeded 0.1 µg/ml for one week post administration, or throughout a 1-year treatment with weekly administrations. After a single administration of atacicept, endogenous BAFF bound to atacicept was detected after 8 h then increased about 100-fold within 2 to 4 weeks. Endogenous heteromers of BAFF and APRIL bound to atacicept also accumulated, but atacicept-APRIL complexes were not detected. In mice receiving intravenous injections of purified complexes pre-formed in vitro, atacicept-BAFF persisted longer (more than a week) than atacicept-APRIL (less than a day). Thus, only biologically inactive BAFF and BAFF-APRIL heteromers accumulate on atacicept in vivo. The measure of active atacicept provides further support for the once-weekly dosing regimen implemented in the clinical development of atacicept.
Collapse
Affiliation(s)
- Mahya Eslami
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Daniela Willen
- Clinical Pharmacology, Translational Medicine, Merck Healthcare KGaA, Darmstadt, Germany
| | - Orestis Papasouliotis
- Translational Medicine, Merck Institute for Pharmacometrics (an affiliate of Merck KGaA), Lausanne, Switzerland
| | | | - Laure Willen
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | | | - Özkan Yalkinoglu
- Clinical Pharmacology, Translational Medicine, Merck Healthcare KGaA, Darmstadt, Germany
| | - Pascal Schneider
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland,*Correspondence: Pascal Schneider,
| |
Collapse
|
7
|
Protein and functional isoform levels and genetic variants of the BAFF and APRIL pathway components in systemic lupus erythematosus. Sci Rep 2022; 12:11219. [PMID: 35780200 PMCID: PMC9250527 DOI: 10.1038/s41598-022-15549-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/24/2022] [Indexed: 11/11/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is the prototype of an autoimmune disease. Belimumab, a monoclonal antibody targets BAFF, is the only biologic approved for SLE and active lupus nephritis. BAFF is a cytokine with a key-regulatory role in the B cell homeostasis, which acts by binding to three receptors: BAFF-R, TACI and BCMA. TACI and BCMA also bind APRIL. Many studies reported elevated soluble BAFF and APRIL levels in the sera of SLE patients, but other questions about the role of this system in the disease remain open. The study aimed to investigate the utility of the cytokine levels in serum and urine as biomarkers, the role of non-functional isoforms, and the association of gene variants with the disease. This case–control study includes a cohort (women, 18–60 years old) of 100 patients (48% with nephritis) and 100 healthy controls. We used ELISA assays to measure the cytokine concentrations in serum (sBAFF and sAPRIL) and urine (uBAFF and uAPRIL); TaqMan Gene Expression Assays to quantify the relative mRNA expression of ΔBAFF, βAPRIL, and εAPRIL, and next-generation sequencing to genotype the cytokine (TNFSF13 and TNFSF13B) and receptor (TNFRSF13B, TNFRSF17 and TNFRSF13C) genes. The statistical tests used were: Kruskal–Wallis (qualitative variables), the Spearman Rho coefficient (correlations), the Chi-square and SKAT (association of common and rare genetic variants, respectively). As expected, sBAFF and sAPRIL levels were higher in patients than in controls (p ≤ 0.001) but found differences between patient subgroups. sBAFF and sAPRIL significantly correlated only in patients with nephritis (rs = 0.67, p ≤ 0.001) and βAPRIL levels were lower in patients with nephritis (p = 0.04), and ΔBAFF levels were lower in patients with dsDNA antibodies (p = 0.04). Rare variants of TNFSF13 and TNFRSF13B and TNFSF13 p.Gly67Arg and TNFRSF13B p.Val220Ala were associated with SLE. Our study supports differences among SLE patient subgroups with diverse clinical features in the BAFF/APRIL pathway. In addition, it suggests the involvement of genetic variants in the susceptibility to the disease.
Collapse
|
8
|
Novel treatment strategies for acetylcholine receptor antibody-positive myasthenia gravis and related disorders. Autoimmun Rev 2022; 21:103104. [PMID: 35452851 DOI: 10.1016/j.autrev.2022.103104] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/18/2022] [Indexed: 11/21/2022]
Abstract
The presence of autoantibodies directed against the muscle nicotinic acetylcholine receptor (AChR) is the most common cause of myasthenia gravis (MG). These antibodies damage the postsynaptic membrane of the neuromuscular junction and cause muscle weakness by depleting AChRs and thus impairing synaptic transmission. As one of the best-characterized antibody-mediated autoimmune diseases, AChR-MG has often served as a reference model for other autoimmune disorders. Classical pharmacological treatments, including broad-spectrum immunosuppressive drugs, are effective in many patients. However, complete remission cannot be achieved in all patients, and 10% of patients do not respond to currently used therapies. This may be attributed to production of autoantibodies by long-lived plasma cells which are resistant to conventional immunosuppressive drugs. Hence, novel therapies specifically targeting plasma cells might be a suitable therapeutic approach for selected patients. Additionally, in order to reduce side effects of broad-spectrum immunosuppression, targeted immunotherapies and symptomatic treatments will be required. This review presents established therapies as well as novel therapeutic approaches for MG and related conditions, with a focus on AChR-MG.
Collapse
|
9
|
Zhang Y, Tian J, Xiao F, Zheng L, Zhu X, Wu L, Zhao C, Wang S, Rui K, Zou H, Lu L. B cell-activating factor and its targeted therapy in autoimmune diseases. Cytokine Growth Factor Rev 2021; 64:57-70. [DOI: 10.1016/j.cytogfr.2021.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/18/2022]
|
10
|
Kulkarni T, Valentine VG, Fei F, Tran-Nguyen TK, Quesada-Arias LD, Mkorombindo T, Pham HP, Simmons SC, Dsouza KG, Luckhardt T, Duncan SR. Correlates of survival after autoantibody reduction therapy for acute IPF exacerbations. PLoS One 2021; 16:e0260345. [PMID: 34813613 PMCID: PMC8610261 DOI: 10.1371/journal.pone.0260345] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 11/08/2021] [Indexed: 11/23/2022] Open
Abstract
Background No medical treatment has proven efficacy for acute exacerbations of idiopathic pulmonary fibrosis (AE-IPF), and this syndrome has a very high mortality. Based on data indicating humoral autoimmune processes are involved in IPF pathogenesis, we treated AE-IPF patients with an autoantibody reduction regimen of therapeutic plasma exchange, rituximab, and intravenous immunoglobulin. This study aimed to identify clinical and autoantibody determinants associated with survival after autoantibody reduction in AE-IPF. Methods Twenty-four(24) AE-IPF patients received the autoantibody reduction regimen. Plasma anti-epithelial autoantibody titers were determined by HEp-2 indirect immunofluorescence assays in 22 patients. Results Mean age of the patients was 70 + 7 years old, and 70% were male. Beneficial clinical responses that occurred early during therapy were a favorable prognostic indicator: supplemental O2 flows needed to maintain resting SaO2>92% significantly decreased and/or walk distances increased among all 10 patients who survived for at least one year. Plasma anti-HEp-2 autoantibody titers were ~-three-fold greater in survivors compared to non-survivors (p<0.02). Anti-HEp-2 titers >1:160 were present in 75% of the evaluable one-year survivors, compared to 29% of non-survivors, and 10 of 12 patients (83%) with anti-HEP-2 titers <1:160 died during the observation period (Hazard Ratio = 3.3, 95% Confidence Interval = 1.02–10.6, p = 0.047). Conclusions Autoantibody reduction therapy is associated with rapid reduction of supplemental oxygen requirements and/or improved ability to ambulate in many AE-IPF patients. Facile anti-epithelial autoantibody assays may help identify those most likely to benefit from these treatments.
Collapse
Affiliation(s)
- Tejaswini Kulkarni
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Vincent G. Valentine
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Fei Fei
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Thi K. Tran-Nguyen
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Luisa D. Quesada-Arias
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States of America
| | - Takudzwa Mkorombindo
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Huy P. Pham
- Department of Pathology, University of Southern California, Los Angeles, CA, United States of America
| | - Sierra C. Simmons
- Department of Pathology, Michigan Pathology Specialists, Spectrum Health Hospitals, Grand Rapids, MI, United States of America
| | - Kevin G. Dsouza
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Tracy Luckhardt
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Steven R. Duncan
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
- * E-mail:
| |
Collapse
|
11
|
Czaja AJ. Review article: targeting the B cell activation system in autoimmune hepatitis. Aliment Pharmacol Ther 2021; 54:902-922. [PMID: 34506662 DOI: 10.1111/apt.16574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/30/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND The B cell activation system, consisting of B cell activating factor and a proliferation-inducing ligand, may have pathogenic effects in autoimmune hepatitis. AIMS To describe the biological actions of the B cell activation system, indicate its possible role in autoimmune diseases, and evaluate its prospects as a therapeutic target in autoimmune hepatitis METHODS: English abstracts were identified in PubMed by multiple search terms. Full length articles were selected for review, and secondary and tertiary bibliographies were developed. RESULTS The B cell activating factor is crucial for the maturation and survival of B cells, and it can co-stimulate T cell activation, proliferation, and survival. It can also modulate the immune response by inducing interleukin 10 production by regulatory B cells. A proliferation-inducing ligand modulates and diversifies the antibody response by inducing class-switch recombination in B cells. It can also increase the proliferation, survival, and antigen activation of T cells. These immune stimulatory actions can be modulated by inducing proliferation of regulatory T cells. The B cell activation system has been implicated in diverse autoimmune diseases, and therapeutic blockade is a management strategy now being evaluated in autoimmune hepatitis. CONCLUSIONS The B cell activation system has profound effects on B and T cell function in autoimmune diseases. Blockade therapy is being actively evaluated in autoimmune hepatitis. Clarification of the critical pathogenic components of the B cell activation system will improve the targeting, efficacy, and safety of blockade therapy in this disease.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| |
Collapse
|
12
|
Baert L, Ahmed MC, Manfroi B, Huard B. The number 13 of the family: a proliferation inducing ligand. Curr Opin Immunol 2021; 71:132-137. [PMID: 34411773 DOI: 10.1016/j.coi.2021.06.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 01/29/2023]
Abstract
The TNF superfamily member a proliferation inducing ligand (APRIL, TNFSF13) plays a late role in humoral immunity at the level of antibody-producing plasmocytes. The recent characterization of the first immunodeficient patient with an inactivating mutation in the APRIL gene provided the last piece of functional data lacking in the human system. Based on this function, APRIL has been considered as a valuable target to dampen unwanted antibody production. After reviewing the late data acquired on the physiological function of APRIL in humoral immunity, we will here review the state of the art regarding APRIL targeting in autoimmune diseases.
Collapse
Affiliation(s)
- Laurie Baert
- Institute for Advanced Biosciences, University Grenoble-Alpes/INSERM U1209/CNRS UMR5309, La Tronche, France
| | - Mashal Claude Ahmed
- Institute for Advanced Biosciences, University Grenoble-Alpes/INSERM U1209/CNRS UMR5309, La Tronche, France
| | - Benoit Manfroi
- Institute for Advanced Biosciences, University Grenoble-Alpes/INSERM U1209/CNRS UMR5309, La Tronche, France
| | - Bertrand Huard
- Institute for Advanced Biosciences, University Grenoble-Alpes/INSERM U1209/CNRS UMR5309, La Tronche, France.
| |
Collapse
|
13
|
Stohl HE, Yu N, Stohl W. First-trimester serum BAFF:sFlt-1 ratio as a candidate early biomarker of spontaneous abortion. Am J Reprod Immunol 2021; 86:e13428. [PMID: 33837577 DOI: 10.1111/aji.13428] [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: 11/23/2020] [Revised: 03/16/2021] [Accepted: 04/06/2021] [Indexed: 11/30/2022] Open
Abstract
PROBLEM Immunologic, angiogenic, and anti-angiogenic factors are associated with spontaneous abortion (SAB). B cell-activating factor (BAFF), a proliferation-inducing ligand (APRIL), placental growth factor (PlGF), and soluble fms-like tyrosine kinase-1 (sFlt-1) may play a role in SAB and may serve singly or in combination as an early biomarker of SAB. METHOD OF STUDY In this prospective observational study, serum sFlt-1, PIGF, BAFF, and APRIL levels were measured in the first trimester of pregnancy in a medically diverse group of women and in non-pregnant controls. Associations and discriminative values of first-trimester sFlt-1, PIGF, BAFF, and APRIL levels and the corresponding APRIL:BAFF, BAFF:sFlt-1, and sFlt-1:PlGF ratios with development of SAB were tested. RESULTS Median serum BAFF level was lower (p = .007) and median serum sFlt-1 level was higher (p < .001), in the first trimester of pregnancy than in non-pregnant controls. SAB developed in 27 of the pregnant women (11.3%), and first-trimester levels of BAFF (but not APRIL) and sFlt-1 (but not PIGF) were associated with SAB. Using optimal cutoffs determined through receiver operating characteristics curves, the best discriminator of SAB was the serum BAFF:sFlt-1 ratio, specifically among non-nulliparous women and women with prior SAB. CONCLUSION First-trimester serum BAFF:sFlt-1 ratio is a candidate indicator/predictor of SAB among non-nulliparous women and women with prior SAB. If validated through additional studies, then early identification of pregnant women at high risk for SAB through this simple blood test would assist in counseling and facilitate clinical trials of therapeutic interventions.
Collapse
Affiliation(s)
- Hindi E Stohl
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ning Yu
- Division of Rheumatology, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - William Stohl
- Division of Rheumatology, Department of Medicine, Los Angeles County + University of Southern California Medical Center, Los Angeles, CA, USA.,Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
14
|
Atrash S, Moyo TK. A Review of Chimeric Antigen Receptor T-Cell Therapy for Myeloma and Lymphoma. Onco Targets Ther 2021; 14:2185-2201. [PMID: 33814917 PMCID: PMC8009535 DOI: 10.2147/ott.s242018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/12/2021] [Indexed: 12/15/2022] Open
Abstract
Collectively, hematological malignancies account for the fourth most common malignancy. Myeloma and lymphoma are the most common types of hematological malignancies. Unfortunately, the management of refractory myeloma and lymphoma remains challenging. The discovery of new immunological therapies, namely chimeric antigen receptors T cells (CAR-T), outlined unprecedented B cell malignancies results. In this context, the CAR-T-based approach has led to the proliferation of many clinical studies. In this review, we will deal with the CAR-T structure, and we will summarize the primary clinical studies assessing the risks and benefits of CAR-T cell therapy. We will also deal with the adverse events and management of cytokine release syndromes/immune effector cell-associated neurotoxicity syndrome (ICANS). Subsequently, we will review potential future improvements to overcome refractoriness and improve expansion while decreasing CAR-T's off-target effects. The advances in the CAR-T platform represent a step forward with promising unlimited future possibilities that made it a paradigm-shifting for the management of B cell malignancies.
Collapse
Affiliation(s)
- Shebli Atrash
- Plasma Cell Disorders Division, Department of Hematologic Oncology & Blood Disorders, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA
| | - Tamara K Moyo
- Lymphoma Division, Department of Hematologic Oncology & Blood Disorders, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA
| |
Collapse
|
15
|
Kim CW, Oh JE, Lee HK. Single Cell Transcriptomic Re-analysis of Immune Cells in Bronchoalveolar Lavage Fluids Reveals the Correlation of B Cell Characteristics and Disease Severity of Patients with SARS-CoV-2 Infection. Immune Netw 2021; 21:e10. [PMID: 33728103 PMCID: PMC7937513 DOI: 10.4110/in.2021.21.e10] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic (severe acute respiratory syndrome coronavirus 2) is a global infectious disease with rapid spread. Some patients have severe symptoms and clinical signs caused by an excessive inflammatory response, which increases the risk of mortality. In this study, we reanalyzed scRNA-seq data of cells from bronchoalveolar lavage fluids of patients with COVID-19 with mild and severe symptoms, focusing on Ab-producing cells. In patients with severe disease, B cells seemed to be more activated and expressed more immunoglobulin genes compared with cells from patients with mild disease, and macrophages expressed higher levels of the TNF superfamily member B-cell activating factor but not of APRIL (a proliferation-inducing ligand). In addition, macrophages from patients with severe disease had increased pro-inflammatory features and pathways associated with Fc receptor-mediated signaling, compared with patients with mild disease. CCR2-positive plasma cells accumulated in patients with severe disease, probably because of increased CCL2 expression on macrophages from patients with severe disease. Together, these results support the hypothesis that different characteristics of B cells might be associated with the severity of COVID-19 infection.
Collapse
Affiliation(s)
- Chae Won Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Ji Eun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea.,The Center for Epidemic Preparedness, KAIST Institute, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
16
|
Eslami M, Meinl E, Eibel H, Willen L, Donzé O, Distl O, Schneider H, Speiser DE, Tsiantoulas D, Yalkinoglu Ö, Samy E, Schneider P. BAFF 60-mer, and Differential BAFF 60-mer Dissociating Activities in Human Serum, Cord Blood and Cerebrospinal Fluid. Front Cell Dev Biol 2020; 8:577662. [PMID: 33240880 PMCID: PMC7677505 DOI: 10.3389/fcell.2020.577662] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
B cell activation factor of the TNF family (BAFF/BLyS), an essential B cell survival factor of which circulating levels are elevated in several autoimmune disorders, is targeted in the clinic for the treatment of systemic lupus erythematosus (SLE). The soluble form of BAFF can exist as 3-mer, or as 60-mer that results from the ordered assembly of twenty 3-mers and that can be obtained from naturally cleaved membrane-bound BAFF or made as a recombinant protein. However, which forms of soluble BAFF exist and act in humans is unclear. In this study, BAFF 3-mer and 60-mer in biological fluids were characterized for size, activity and response to specific stimulators or inhibitors of BAFF. Human cerebrospinal fluids (CSF) from patients with multiple sclerosis and adult human sera contained exclusively BAFF 3-mer in these assays, also when BAFF concentrations were moderately SLE or highly (BAFFR-deficient individual) increased. Human sera, but not CSF, contained a high molecular weight, saturable activity that dissociated preformed recombinant BAFF 60-mer into 3-mer. This activity was lower in cord blood. Cord blood displayed BAFF levels 10-fold higher than in adults and consistently contained a fair proportion of active high molecular weight BAFF able to dissociate into 3-mer but not endowed with all properties of recombinant BAFF 60-mer. If BAFF 60-mer is produced in humans, it is dissociated, or at least attenuated in the circulation.
Collapse
Affiliation(s)
- Mahya Eslami
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital of the Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hermann Eibel
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Freiburg, Germany
| | - Laure Willen
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | - Ottmar Distl
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Holm Schneider
- Department of Pediatrics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Daniel E Speiser
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | | | - Özkan Yalkinoglu
- Clinical Pharmacology, Quantitative Pharmacology, Translational Medicine, Merck KGaA, Darmstadt, Germany
| | - Eileen Samy
- Business of Merck KGaA, EMD Serono Research & Development Institute, Inc., Billerica, MA, United States
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
17
|
Magliozzi R, Marastoni D, Calabrese M. The BAFF / APRIL system as therapeutic target in multiple sclerosis. Expert Opin Ther Targets 2020; 24:1135-1145. [PMID: 32900236 DOI: 10.1080/14728222.2020.1821647] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The complex system of BAFF (B-cell-activating factor of the TNF family) and APRIL (A proliferation-inducing ligand) has been studied in animal models of autoimmune diseases such as those resembling human systemic lupus erythematosus and Sjogren's syndrome and multiple sclerosis (MS). Accumulating evidence suggests that BAFF and APRIL have a physiological role in B cell immunity regulation, however inappropriate production of these factors may represent a key event which disrupts immune tolerance which is associated with systemic autoimmune diseases. AREAS COVERED We provide an update on the latest studies of the BAFF/APRIL system in multiple sclerosis, as well as on related clinical trials. EXPERT OPINION Experimental and clinical evidence suggests that increased BAFF levels may interfere directly and indirectly with B cell immunity; this can lead to breakdown of immune tolerance, the production of autoantibodies and continuous local intracerebral inflammation and brain tissue destruction. A more comprehensive understanding of the cell/molecular mechanism immune reactions specifically regulated by BAFF/APRIL in MS would better elucidate the specific cell phenotype targeted by actual anti-BAFF/APRIL therapies; this may enable the identification of either specific biomarkers of MS subgroups that would benefit of anti-BAFF/APRIL treatments or new targets of MS-specific anti-BAFF/APRIL therapies.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona , Verona, Italy
| | - Damiano Marastoni
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona , Verona, Italy
| | - Massimiliano Calabrese
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona , Verona, Italy
| |
Collapse
|
18
|
Willen D, Uhl W, Wolna P, Papasouliotis O, Yalkinoglu Ö. Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of Atacicept in a Randomized Trial in Healthy Caucasian and Japanese Subjects. Eur J Drug Metab Pharmacokinet 2020; 45:27-40. [PMID: 31529406 PMCID: PMC6994531 DOI: 10.1007/s13318-019-00575-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Background and Objective Atacicept is an inhibitor of the B lymphocyte stimulator (BLyS) and a proliferation-inducing ligand (APRIL), and is being studied in relation to immunological disease. Currently, limited data on atacicept are available in non-Caucasian subjects. Pharmacokinetic data from earlier studies of atacicept were derived using an enzyme-linked immunosorbent assay (ELISA), which was subsequently found to have inadequacies. Hence, a new bioanalytical ELISA for total atacicept was developed and validated. We conducted this randomized, double-blind, placebo-controlled phase I study to compare the safety, tolerability, pharmacokinetics, and pharmacodynamics of atacicept in healthy Japanese and Caucasian subjects while generating pharmacokinetic data using the new ELISA. Methods Japanese subjects aged ≥ 18 to ≤ 55 years (n = 24) were randomized (1:1:1:1) to a single subcutaneous dose of atacicept 25, 75, or 150 mg or placebo. Caucasian subjects were then enrolled to match the Japanese subjects’ gender, body weight (± 20%), and height (± 15%). Results Atacicept was well tolerated and there were no clinically significant differences in treatment-emergent adverse events (TEAEs), vital signs, or laboratory parameters between the Japanese and Caucasian subjects. Most (90%) TEAEs were mild; no severe or serious TEAEs or deaths occurred. Weight-adjusted atacicept exposure was comparable between ethnicities and across doses: the Japanese/Caucasian ratio of the area under the serum concentration–time curve from time zero to the last sampling point (AUC0–t) was 107.21% (90% CI 93.42–123.02%) and the Japanese/Caucasian ratio of maximum serum concentration (Cmax) was 95.74% (90% CI 74.26–123.43%; ANCOVA). Median time to reach Cmax (tmax) was 20–60 h across all subjects. Dose–exposure relationships were comparable for the two ethnicities, with dose-normalized AUC0–t decreasing with increasing dose, indicating nonlinear pharmacokinetics for the doses examined. There were no statistically significant differences between ethnicities in the pharmacokinetics–dose relationship. Some transient dose-related decreases in mean serum immunoglobulin (Ig)A and IgM, but not IgG, were observed after atacicept administration. There were small transient increases in peripheral B cell numbers in the first 4 days after dosing that were larger with atacicept than with placebo, with no apparent dose relationship. No anti-atacicept antibodies were detected. Conclusion The safety, pharmacokinetic, and pharmacodynamic profiles of atacicept in healthy Japanese subjects were comparable to those in healthy Caucasian subjects. EudraCT-ID: 2013-002703-34. Electronic supplementary material The online version of this article (10.1007/s13318-019-00575-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniela Willen
- Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Wolfgang Uhl
- Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Peter Wolna
- Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Orestis Papasouliotis
- Merck Institute for Pharmacometrics (An Affiliate of Merck KGaA, Darmstadt, Germany), Lausanne, Switzerland
| | | |
Collapse
|
19
|
Liu Y, Li X, Han Y, Qiu Z, Song X, Li B, Zhang H, Wang H, Feng K, Liu L, Wang J, Sun M, Li T. High APRIL Levels Are Associated With Slow Disease Progression and Low Immune Activation in Chronic HIV-1-Infected Patients. Front Med (Lausanne) 2020; 7:299. [PMID: 32850873 PMCID: PMC7396611 DOI: 10.3389/fmed.2020.00299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Objective: B-cell-activating factor (BAFF) has been determined to be involved in HIV-1 infection and is correlated with disease progression, while its homologous molecule, a proliferation-inducing ligand (APRIL), is less frequently reported, and its role remains unclear. We aimed to characterize the APRIL levels in subjects with different HIV-1 infection statuses and determine the relationships with disease progression and immune activation. Methods: The plasma levels of APRIL were compared among 17 long-term non-progressors (LTNPs), 17 typical progressors (TPs), 10 ART-treated patients, and 10 healthy donors (HDs). Seventeen LTNPs and a subset of TPs (n = 6) who initiated ART were assessed longitudinally. The correlations between the APRIL levels and markers of disease progression, B-cell count and specific antibody response, and markers of immune activation and functional cells were analyzed. Results: The circulating APRIL levels were significantly elevated in the LTNPs relative to the TPs, ART-treated patients, and HDs. The longitudinal investigation revealed that the APRIL levels were decreased during follow-up in the LTNPs. ART did not significantly influence the APRIL levels. The levels of plasma APRIL were negatively correlated with the plasma HIV-1 viral load and cellular HIV-1 DNA levels and positively correlated with the CD4+ T-cell count and CD4/CD8 ratio. An inverse correlation was observed between the APRIL and BAFF levels. Furthermore, the APRIL levels were negatively correlated with the frequency of activated CD8+ T cells and levels of interferon gamma-induced protein 10 (IP-10) and monocyte chemoattractant protein-1 (MCP-1). Finally, positive correlations were observed among the APRIL levels, the frequency of CD8+CD28+ T cells, and natural killer (NK) cell count. Conclusion: The APRIL levels were elevated in the LTNPs and negatively correlated with disease progression and immune activation, suggesting likely protective activity in HIV-1 infection.
Collapse
Affiliation(s)
- Yubin Liu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiuxia Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Han
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhifeng Qiu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaojing Song
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Bingxiang Li
- Institute of Medical Biology, Peking Union Medical College and Chinese Academy of Medical Sciences, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| | - Han Zhang
- Institute of Medical Biology, Peking Union Medical College and Chinese Academy of Medical Sciences, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| | - Hongye Wang
- Institute of Medical Biology, Peking Union Medical College and Chinese Academy of Medical Sciences, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| | - Kai Feng
- Institute of Medical Biology, Peking Union Medical College and Chinese Academy of Medical Sciences, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| | - Longding Liu
- Institute of Medical Biology, Peking Union Medical College and Chinese Academy of Medical Sciences, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| | - Jingjing Wang
- Institute of Medical Biology, Peking Union Medical College and Chinese Academy of Medical Sciences, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| | - Ming Sun
- Institute of Medical Biology, Peking Union Medical College and Chinese Academy of Medical Sciences, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| | - Taisheng Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing, China.,School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
20
|
Salazar-Camarena DC, Palafox-Sánchez CA, Cruz A, Marín-Rosales M, Muñoz-Valle JF. Analysis of the receptor BCMA as a biomarker in systemic lupus erythematosus patients. Sci Rep 2020; 10:6236. [PMID: 32277232 PMCID: PMC7148319 DOI: 10.1038/s41598-020-63390-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 03/30/2020] [Indexed: 11/08/2022] Open
Abstract
B cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL) play central roles in B cell development and maturation. Soluble forms of their receptors can be generated by proteolytic cleavage; however, their physiological and clinical roles are unknown. This study aimed to assess the relationships between the receptor soluble B cell maturation antigen (sBCMA) and clinical variables in systemic lupus erythematosus (SLE) patients. Serum cytokine concentrations were measured by ELISA for 129 SLE patients and 34 healthy controls (HCs), and the expression of the receptor BCMA was evaluated on B and plasma cells from 40 subjects. SLE patients showed aberrant expression of the receptor BCMA on B and plasma cells. Soluble levels of the receptor sBCMA and its ligands sAPRIL and sBAFF were increased in SLE patients compared with HCs. Additionally, sBCMA (rs = 0.6177) and sAPRIL (rs = 0.4952) correlated strongly with disease activity. Active SLE patients who achieved low disease activity showed decreased sBCMA (53.30 vs 35.30 ng/mL; p < 0.05) and sBAFF (4.48 vs 2.27 ng/mL; p < 0.05) serum levels after treatment, while sAPRIL expression remained unchanged. At a cutoff value of 22.40 ng/mL, sAPRIL showed high sensitivity (96.12%) and specificity (94.12%) for discrimination between HCs and SLE patients, while sBAFF showed lower sensitivity (82.2%) but higher specificity (94.1%) at a cutoff of 1.195 ng/mL. Relatively high levels of sAPRIL and sBCMA clustered active SLE patients. The receptor sBCMA could be a potential biomarker of disease activity in SLE.
Collapse
Affiliation(s)
- Diana Celeste Salazar-Camarena
- Research Institute in Biomedical Sciences, University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Claudia Azucena Palafox-Sánchez
- Research Institute in Biomedical Sciences, University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico.
| | - Alvaro Cruz
- Research Institute in Biomedical Sciences, University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Miguel Marín-Rosales
- Department of Rheumatology, West Medical Hospital, Ministry of Health, Zapopan, Mexico
| | - José Francisco Muñoz-Valle
- Research Institute in Biomedical Sciences, University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico
| |
Collapse
|
21
|
Chang HD, Tokoyoda K, Hoyer B, Alexander T, Khodadadi L, Mei H, Dörner T, Hiepe F, Burmester GR, Radbruch A. Pathogenic memory plasma cells in autoimmunity. Curr Opin Immunol 2019; 61:86-91. [PMID: 31675681 PMCID: PMC6908965 DOI: 10.1016/j.coi.2019.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/09/2019] [Accepted: 09/22/2019] [Indexed: 01/06/2023]
Abstract
Memory plasma cells are long-lived but require specialized niches for their survival. Memory plasma cells are refractory to conventional immunosuppression. Pathogenic memory plasma cells represent ‘roadblocks’ to response to conventional therapy. Strategies for (selective) targeting of memory plasma cells are in preclinical and clinical tests.
Collapse
Affiliation(s)
- Hyun-Dong Chang
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Charitéplatz 1, 10117 Berlin, Germany
| | - Koji Tokoyoda
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Charitéplatz 1, 10117 Berlin, Germany
| | - Bimba Hoyer
- Universitätsklinikum Schleswig-Holstein, Clinic for Internal Medicine I, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Tobias Alexander
- Charité Universitätsmedizin Berlin, Department of Rheumatology and Clinical Immunology, Charitéplatz 1, 10117 Berlin, Germany
| | - Laleh Khodadadi
- Charité Universitätsmedizin Berlin, Department of Rheumatology and Clinical Immunology, Charitéplatz 1, 10117 Berlin, Germany
| | - Henrik Mei
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Charitéplatz 1, 10117 Berlin, Germany
| | - Thomas Dörner
- Charité Universitätsmedizin Berlin, Department of Rheumatology and Clinical Immunology, Charitéplatz 1, 10117 Berlin, Germany
| | - Falk Hiepe
- Charité Universitätsmedizin Berlin, Department of Rheumatology and Clinical Immunology, Charitéplatz 1, 10117 Berlin, Germany
| | - Gerd-Rüdiger Burmester
- Charité Universitätsmedizin Berlin, Department of Rheumatology and Clinical Immunology, Charitéplatz 1, 10117 Berlin, Germany
| | - Andreas Radbruch
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute (DRFZ), Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
22
|
Gordon C, Bassi R, Chang P, Kao A, Jayne D, Wofsy D, Fleuranceau-Morel P. Integrated safety profile of atacicept: an analysis of pooled data from the atacicept clinical trial programme. Rheumatol Adv Pract 2019; 3:rkz021. [PMID: 31528843 PMCID: PMC6735746 DOI: 10.1093/rap/rkz021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 07/11/2019] [Indexed: 11/17/2022] Open
Abstract
Objective To characterize the overall safety profile of atacicept, we conducted an integrated analysis of pooled safety data from all 17 clinical studies to date. Methods Three data sets were used to investigate safety endpoints: a double-blind placebo-controlled set (n = 1568), an SLE set (n = 761) and a full analysis set (n = 1845; including all 17 studies). Results Of 1568 patients in the double-blind placebo-controlled-set, 30.8% received placebo, and 8.2, 24.5 and 36.5% received atacicept 25, 75 and 150 mg, respectively. Treatment-emergent adverse event (TEAE) rates (adjusted by treatment-exposure) were generally higher with atacicept vs placebo, but no consistent association was found between atacicept dose and specific TEAEs or mortality. Serious infection and serious TEAE rates were similar for atacicept and placebo. The TEAE-related discontinuation rates were higher with atacicept vs placebo (16.1 vs 10.9/100 patient-years). In the full analysis set, 11 deaths occurred during treatment. Across indications, exposure-adjusted mortality rates/100 patient-years (95% CI) were 3.60 (0.90, 14.38), 0.34 (0.05, 2.43) and 1.18 (0.49, 2.82) with atacicept 25, 75 and 150 mg, respectively, and 0.44 (0.06, 3.12) with placebo. In SLE patients, exposure-adjusted mortality rates were 1.45 (0.54, 3.87) with atacicept 150 mg and 0.78 (0.29, 2.07) across all atacicept-treated patients. No deaths occurred with atacicept 75 mg or placebo. In the SLE and double-blind placebo-controlled sets, pharmacodynamic effects of atacicept were not associated with increased infection rates. Conclusion The results of this integrated safety analysis support further development and evaluation of atacicept in selected patients for whom potential benefits might outweigh risks.
Collapse
Affiliation(s)
- Caroline Gordon
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Roberto Bassi
- EMD Serono Research & Development Institute, Inc. (A Business of Merck KGaA, Darmstadt, Germany), Billerica, MA, USA
| | - Peter Chang
- EMD Serono Research & Development Institute, Inc. (A Business of Merck KGaA, Darmstadt, Germany), Billerica, MA, USA
| | - Amy Kao
- EMD Serono Research & Development Institute, Inc. (A Business of Merck KGaA, Darmstadt, Germany), Billerica, MA, USA
| | - David Jayne
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - David Wofsy
- Russell/Engleman Rheumatology Research Center, University of California, San Francisco, CA, USA
| | - Patricia Fleuranceau-Morel
- EMD Serono Research & Development Institute, Inc. (A Business of Merck KGaA, Darmstadt, Germany), Billerica, MA, USA
| |
Collapse
|
23
|
The role of APRIL - A proliferation inducing ligand - In autoimmune diseases and expectations from its targeting. J Autoimmun 2018; 95:179-190. [DOI: 10.1016/j.jaut.2018.10.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
|
24
|
Rodríguez-Carrio J, Alperi-López M, López P, Ballina-García FJ, Suárez A. Profiling of B-Cell Factors and Their Decoy Receptors in Rheumatoid Arthritis: Association With Clinical Features and Treatment Outcomes. Front Immunol 2018; 9:2351. [PMID: 30369929 PMCID: PMC6194314 DOI: 10.3389/fimmu.2018.02351] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/24/2018] [Indexed: 12/27/2022] Open
Abstract
Introduction: B-cell activation is pivotal in rheumatoid arthritis (RA) pathogenesis and represents a relevant therapeutic target. The main aim of this study was to characterize the profiles of B-cell factors and their decoy receptors in RA and evaluate their clinical relevance. Methods: sBLyS, sAPRIL, sBCMA, sTACI, sBLyS-R, and several cytokines' serum levels were measured by immunoassays in 104 RA patients and 33 healthy controls (HC). An additional group of 42 systemic lupus erythematosus (SLE) patients were enrolled as disease controls. Whole blood IFI44, IFI44L, IFI6, and MX1 gene expression was measured and averaged into an IFN-score. BLyS membrane expression (mBLyS) was assessed on blood cell subsets by flow cytometry. Results: increased sAPRIL and sBCMA levels were found in RA, whereas BLyS was elevated in very early RA (VERA). No differences were observed for sTACI and sBLyS-R. An increased sBLyS/sBLyS-R ratio was associated with poor clinical outcome at 6 and 12 months in VERA, whereas a positive association with disease activity was observed in established disease. Increased mBLyS expression was found on monocytes, mDCs, neutrophils and B-cells in RA, to a similar extent that in SLE patients. Cluster analysis identified a specific B-cell factors profile overrepresented in RA and associated with autoantibodies, elevated proinflammatory cytokines (IFNα, MIP1α, TNFα, IL-37, and GM-CSF) and increased type-I IFN signature. Increasing sBCMA and sBLyS serum levels upon treatment and mBLyS expression at baseline on monocytes and mDCs, but not B-cells, were associated with poor clinical outcome upon TNFα-blockade. Conclusions: profound and complex alterations of soluble and membrane-bound B-cell factors are observed in RA associated with clinical outcomes, thus supporting its applicability to guide patient stratification along disease course.
Collapse
Affiliation(s)
- Javier Rodríguez-Carrio
- Area of Immunology, Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Mercedes Alperi-López
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Department of Rheumatology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Patricia López
- Area of Immunology, Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Francisco J Ballina-García
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Department of Rheumatology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Ana Suárez
- Area of Immunology, Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| |
Collapse
|
25
|
Samy E, Wax S, Huard B, Hess H, Schneider P. Targeting BAFF and APRIL in systemic lupus erythematosus and other antibody-associated diseases. Int Rev Immunol 2017; 36:3-19. [PMID: 28215100 DOI: 10.1080/08830185.2016.1276903] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The B cell-stimulating molecules, BAFF (B cell activating factor) and APRIL (a proliferation-inducing ligand), are critical factors in the maintenance of the B cell pool and humoral immunity. In addition, BAFF and APRIL are involved in the pathogenesis of a number of human autoimmune diseases, with elevated levels of these cytokines detected in the sera of patients with systemic lupus erythematosus (SLE), IgA nephropathy, Sjögren's syndrome, and rheumatoid arthritis. As such, both molecules are rational targets for new therapies in B cell-driven autoimmune diseases, and several inhibitors of BAFF or BAFF and APRIL together have been investigated in clinical trials. These include the BAFF/APRIL dual inhibitor, atacicept, and the BAFF inhibitor, belimumab, which is approved as an add-on therapy for patients with active SLE. Post hoc analyses of these trials indicate that baseline serum levels of BAFF and BAFF/APRIL correlate with treatment response to belimumab and atacicept, respectively, suggesting a role for the two molecules as predictive biomarkers. It will, however, be important to refine future testing to identify active forms of BAFF and APRIL in the circulation, as well as to distinguish between homotrimer and heteromer configurations. In this review, we discuss the rationale for dual BAFF/APRIL inhibition versus single BAFF inhibition in autoimmune disease, by focusing on the similarities and differences between the physiological and pathogenic roles of the two molecules. A summary of the preclinical and clinical data currently available is also presented.
Collapse
Affiliation(s)
- Eileen Samy
- a EMD Serono Research & Development Institute, Inc. , Billerica , Massachusetts , USA
| | - Stephen Wax
- a EMD Serono Research & Development Institute, Inc. , Billerica , Massachusetts , USA
| | - Bertrand Huard
- b Institute for Advanced Biosciences , University Grenoble Alpes , INSERM U1209, Grenoble , France
| | | | - Pascal Schneider
- d Department of Biochemistry , University of Lausanne , Lausanne , Switzerland
| |
Collapse
|
26
|
Davis LS, Reimold AM. Research and therapeutics-traditional and emerging therapies in systemic lupus erythematosus. Rheumatology (Oxford) 2017; 56:i100-i113. [PMID: 28375452 DOI: 10.1093/rheumatology/kew417] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Indexed: 12/21/2022] Open
Abstract
This review summarizes traditional and emerging therapies for SLE. Evidence suggests that the heterogeneity of SLE is a crucial aspect contributing to the failure of large clinical trials for new targeted therapies. A clearer understanding of the mechanisms driving disease pathogenesis combined with recent advances in medical science are predicted to enable accelerated progress towards improved SLE diagnosis and personalized approaches to treatment.
Collapse
Affiliation(s)
- Laurie S Davis
- Rheumatic Diseases Division, Department of Internal Medicine, University of Texas Southwestern Medical Center
| | - Andreas M Reimold
- Rheumatic Diseases Division, Department of Internal Medicine, University of Texas Southwestern Medical Center.,Dallas VA Medical Center, Dallas, TX, USA
| |
Collapse
|
27
|
Hoyne GF, Elliott H, Mutsaers SE, Prêle CM. Idiopathic pulmonary fibrosis and a role for autoimmunity. Immunol Cell Biol 2017; 95:577-583. [DOI: 10.1038/icb.2017.22] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Gerard F Hoyne
- School of Health Sciences, University of Notre Dame Australia Fremantle Western Australia Australia
- Institute of Health Research, University of Notre Dame Fremantle Western Australia Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, Harry Perkins Institute of Medical Research, University of Western Australia Nedlands Western Australia Australia
| | - Hannah Elliott
- School of Health Sciences, University of Notre Dame Australia Fremantle Western Australia Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, Harry Perkins Institute of Medical Research, University of Western Australia Nedlands Western Australia Australia
| | - Steven E Mutsaers
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, Harry Perkins Institute of Medical Research, University of Western Australia Nedlands Western Australia Australia
- Institute for Respiratory Health, Centre for Respiratory Health, School of Medicine and Pharmacology, University of Western Australia Nedlands Western Australia Australia
| | - Cecilia M Prêle
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, Harry Perkins Institute of Medical Research, University of Western Australia Nedlands Western Australia Australia
- Institute for Respiratory Health, Centre for Respiratory Health, School of Medicine and Pharmacology, University of Western Australia Nedlands Western Australia Australia
| |
Collapse
|
28
|
Haselmayer P, Vigolo M, Nys J, Schneider P, Hess H. A mouse model of systemic lupus erythematosus responds better to soluble TACI than to soluble BAFFR, correlating with depletion of plasma cells. Eur J Immunol 2017; 47:1075-1085. [PMID: 28383107 PMCID: PMC5518279 DOI: 10.1002/eji.201746934] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/16/2017] [Accepted: 03/30/2017] [Indexed: 11/05/2022]
Abstract
The TNF family cytokines B-cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL) support plasma cell survival. It is known that inhibitors of BAFF only (BAFFR-Fc) or BAFF and APRIL (TACI-Fc) administered early enough in an NZB/NZW F1 mouse model of systemic lupus erythematosus (SLE) ameliorate clinical outcomes, pointing to a pathogenic role of BAFF. In the present study, TACI-Fc administrated at a later stage of disease, after onset of autoimmunity, decreased the number of bone marrow plasma cells and slowed down further formation of autoantibodies. TACI-Fc prevented renal damage during a 12-week treatment period regardless of autoantibody levels, while BAFFR-Fc did not despite a similar BAFF-blocking activity in vivo. TACI-Fc also decreased established plasma cells in a T-dependent hapten/carrier immunization system better than single inhibitors of BAFF or APRIL, and sometimes better than combined single inhibitors with at least equivalent BAFF and APRIL inhibitory activities. These results indicate that TACI-Fc can prevent symptoms of renal damage in a mouse model of SLE when BAFFR-Fc cannot, and point to a plasticity of plasma cells for survival factors. Targeting plasma cells with TACI-Fc might be beneficial to prevent autoantibody-mediated damages in SLE.
Collapse
Affiliation(s)
- Philipp Haselmayer
- Department of Immunopharmacology, Immunology Translational Innovation Platform, Merck KGaA, Darmstadt, Germany
| | - Michele Vigolo
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Josquin Nys
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Henry Hess
- Department of Immunopharmacology, Immunology Translational Innovation Platform, Merck KGaA, Darmstadt, Germany
| |
Collapse
|
29
|
Tran NL, Schneider P, Santiago-Raber ML. TACI-dependent APRIL signaling maintains autoreactive B cells in a mouse model of systemic lupus erythematosus. Eur J Immunol 2017; 47:713-723. [DOI: 10.1002/eji.201646630] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 01/16/2017] [Accepted: 03/01/2017] [Indexed: 11/05/2022]
Affiliation(s)
- Ngoc Lan Tran
- Department of Pathology and Immunology; Faculty of Medicine; University of Geneva; Geneva Switzerland
| | - Pascal Schneider
- Department of Biochemistry; University of Lausanne; Epalinges Switzerland
| | | |
Collapse
|
30
|
Lenert A, Niewold TB, Lenert P. Spotlight on blisibimod and its potential in the treatment of systemic lupus erythematosus: evidence to date. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:747-757. [PMID: 28331294 PMCID: PMC5357079 DOI: 10.2147/dddt.s114552] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
B cells in general and BAFF (B cell activating factor of the tumor necrosis factor [TNF] family) in particular have been primary targets of recent clinical trials in systemic lupus erythematosus (SLE). In 2011, belimumab, a monoclonal antibody against BAFF, became the first biologic agent approved for the treatment of SLE. Follow-up studies have shown excellent long-term safety and tolerability of belimumab. In this review, we critically analyze blisibimod, a novel BAFF-neutralizing agent. In contrast to belimumab that only blocks soluble BAFF trimer but not soluble 60-mer or membrane BAFF, blisibimod blocks with high affinity all three forms of BAFF. Furthermore, blisibimod has a unique structure built on four high-affinity BAFF-binding peptides fused to the IgG1-Fc carrier. It was tested in phase I and II trials in SLE where it showed safety and tolerability. While it failed to reach the primary endpoint in a recent phase II trial, post hoc analysis demonstrated its efficacy in SLE patients with higher disease activity. Based on these results, blisibimod is currently undergoing phase III trials targeting this responder subpopulation of SLE patients. The advantage of blisibimod, compared to its competitors, lies in its higher avidity for BAFF, but a possible drawback may come from its immunogenic potential and the anticipated loss of efficacy over time.
Collapse
Affiliation(s)
- Aleksander Lenert
- Division of Rheumatology, University of Kentucky, Kentucky Clinic, Lexington, KY
| | - Timothy B Niewold
- Division of Rheumatology and Department of Immunology, Mayo Clinic, Rochester, MN
| | - Petar Lenert
- Division of Immunology, Department of Internal Medicine, The University of Iowa, Iowa City, IA, USA
| |
Collapse
|
31
|
Stohl W. Inhibition of B cell activating factor (BAFF) in the management of systemic lupus erythematosus (SLE). Expert Rev Clin Immunol 2017; 13:623-633. [DOI: 10.1080/1744666x.2017.1291343] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- William Stohl
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
32
|
Stohl W. The Future of B-cell Activating Factor Antagonists in the Treatment of Systemic Lupus Erythematosus. JOURNAL OF RHEUMATIC DISEASES 2017. [DOI: 10.4078/jrd.2017.24.2.65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- William Stohl
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
33
|
Clancy RM, Markham AJ, Buyon JP. Endosomal Toll-like receptors in clinically overt and silent autoimmunity. Immunol Rev 2016; 269:76-84. [PMID: 26683146 DOI: 10.1111/imr.12383] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Toll-like receptors (TLRs), first identified as pattern recognition receptors, are now recognized to serve as a key interface between innate and adaptive immunity. Systemic lupus erythematosus (SLE) is characterized by both continuous and cyclic stimulation of the innate and adaptive immune system by endogenous nucleic acids released from apoptotic or necrotic cells. TLR7 and TLR9 function as innate sensors of viral infection as their ligands are ssRNA and dsDNA, respectively. Recognition of self nucleic acids by endosomal TLRs in B cells and pDCs is thought to be an important step in the pathogenesis of SLE, generating anti-nuclear antibodies and producing type I IFN. In this review, we take a specific look at how TLR7, non-coding RNA, and SSA/Ro60 can contribute to clinical autoimmunity and organ damage in the context of neonatal lupus (NL). Although 15 times less common than SLE, NL provides a unique opportunity to study two different aspects of autoimmunity: passively acquired tissue injury in a developing fetus and clinical progression of disease in an asymptomatic mother found to have anti-Ro60 autoantibodies only after identification of heart block/rash in a child. Finally, we discuss hydroxychloroquine (HCQ) use by asymptomatic subjects which may forestall the clinical expression of autoimmunity.
Collapse
Affiliation(s)
- Robert M Clancy
- Division of Rheumatology, Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - Androo J Markham
- Division of Rheumatology, Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - Jill P Buyon
- Division of Rheumatology, Department of Medicine, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
34
|
B Cell-Activating Factor as a New Potential Marker in Inflammatory Bowel Disease. Dig Dis Sci 2016; 61:2608-18. [PMID: 27056038 DOI: 10.1007/s10620-016-4136-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 03/17/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND B cell-activating factor (BAFF) has been proposed to be a regulator of B cell and T cell immune responses and be associated with inflammatory processes in autoimmunity and B cell malignancies. No study has reported the role of BAFF in inflammatory bowel disease (IBD). AIMS The purpose of this study was to investigate expression and concentrations of BAFF in IBD and determine its value to discriminate patients with IBD. METHODS Seventy-eight ulcerative colitis (UC) patients, 37 Crohn's disease (CD) patients, 12 irritable bowel syndrome (IBS) patients and 44 healthy controls were recruited. We examined serum and faecal BAFF levels using enzyme-linked immunosorbent assay. Intestinal BAFF expression was analysed in biopsies obtained from IBD patients. Intestinal mucosa localization of BAFF was conducted by immunofluorescence. RESULTS The median (25th-75th percentile) serum BAFF concentration (pg/ml) was 1430 (1105-1624) in CD patients, 1472 (1018-1772) in UC patients and 977 (482-1345) in healthy controls. Serum BAFF was 64 % sensitive and 93 % specific for identifying active IBD from healthy controls. The BAFF expression was significantly increased in biopsy specimens from IBD patients. Fecal BAFF concentration was 369 (326-493) pg/ml in CD patients, 542 (358-1758) pg/ml in UC patients, 294 (287-299) pg/ml in IBS patients and 295 (284-309) pg/ml in healthy controls. Fecal BAFF was 90 % sensitive and 96 % specific for identifying active IBD from healthy controls and IBS patients. CONCLUSION The novel association between BAFF and IBD seems to identify that BAFF might regulate the inflammatory process in these diseases and it appears to be a potential marker of IBD.
Collapse
|
35
|
Hou W, Meng X, Wang Y, Mo W, Wu Y, Yu M. Characterization and high-yield production of non- N-glycosylated recombinant human BCMA-Fc in Pichia pastoris. Eng Life Sci 2016; 17:96-106. [PMID: 32624756 DOI: 10.1002/elsc.201600039] [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: 01/29/2016] [Revised: 05/17/2016] [Accepted: 06/09/2016] [Indexed: 11/11/2022] Open
Abstract
B-cell maturation antigen (BCMA) fused at the C-terminus to the Fc portion of human IgG1 (BCMA-Fc) blocks B-cell activating factor (BAFF) and proliferation-inducing ligand (APRIL)-mediated B-cell activation, leading to immune disorders. The fusion protein has been cloned and produced by several engineering cell lines. To reduce cost and enhance production, we attempted to express recombinant human BCMA-Fc (rhBCMA-Fc) in Pichia pastoris under the control of the AOX1 methanol-inducible promoter. To produce the target protein with uniform molecular weight and reduced immunogenicity, we mutated two predicted N-linked glycosylation sites. The secretory yield was improved by codon optimization of the target gene sequence. After fed-batch fermentation under optimized conditions, the highest yield (207 mg/L) of rhBCMA-Fc was obtained with high productivity (3.45 mg/L/h). The purified functional rhBCMA-Fc possessed high-binding affinity to APRIL and dose-dependent inhibition of APRIL-induced proliferative activity in vitro through three-step purification. Thus, this yeast-derived expression method could be a low-cost and effective alternative to the production of rhBCMA-Fc in mammalian cell lines.
Collapse
Affiliation(s)
- Weihua Hou
- Ministry of Education and Department of Biochemistry and Molecular Biology School of Basic Medicine Fudan University Shanghai China
| | - Xianchao Meng
- Ministry of Education and Department of Biochemistry and Molecular Biology School of Basic Medicine Fudan University Shanghai China
| | - Yuxiong Wang
- Ministry of Education and Department of Biochemistry and Molecular Biology School of Basic Medicine Fudan University Shanghai China
| | - Wei Mo
- Ministry of Education and Department of Biochemistry and Molecular Biology School of Basic Medicine Fudan University Shanghai China
| | - Yi Wu
- Ministry of Education and Department of Biochemistry and Molecular Biology School of Basic Medicine Fudan University Shanghai China
| | - Min Yu
- Ministry of Education and Department of Biochemistry and Molecular Biology School of Basic Medicine Fudan University Shanghai China
| |
Collapse
|
36
|
Polverino F, Laucho-Contreras M, Rojas Quintero J, Divo M, Pinto-Plata V, Sholl L, de-Torres JP, Celli BR, Owen CA. Increased expression of A Proliferation-inducing Ligand (APRIL) in lung leukocytes and alveolar epithelial cells in COPD patients with non small cell lung cancer: a possible link between COPD and lung cancer? Multidiscip Respir Med 2016; 11:17. [PMID: 27047662 PMCID: PMC4819280 DOI: 10.1186/s40248-016-0051-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 02/17/2016] [Indexed: 11/26/2022] Open
Abstract
Background Chronic Obstructive Pulmonary Disease (COPD) is characterized by an excessive activation of the adaptive immune system and, in particular, uncontrolled expansion of the B-cell pool. One of the key promoters of B cell expansion is A PRoliferation-Inducing Ligand (APRIL). APRIL has been strongly linked to non small cell lung cancer (NSCLC) onset and progression previously. However, little is known about the expression of APRIL in the lungs of COPD patients. Methods Using immuno-fluorescence staining, the expression of APRIL was assessed in sections of lungs from 4 subjects with primary diagnosis of COPD (FEV1 33 ± 20 % predicted), 4 subjects with primary diagnosis of NSCLC, 4 subjects diagnosed with both COPD and NSCLC, smokers without COPD or NSCLC and 3 healthy never-smokers. The percentage of B cells, alveolar macrophages (AMs) and polymorphonuclear neutrophils (PMNs) in the lung and alveolar epithelial cells (AECs) that stained positively for APRIL was quantified using epi-fluorescence microscopy and image analysis software. Results The percentage of APRIL-expressing B cells, AMs, PMNs and alveolar epithelial cells (AECs) was higher in patients having both COPD and NSCLC than in patients with either COPD or NSCLC alone, SC or NSC (p < 0.03 for all comparisons). The percentage of APRIL-expressing AMs and AECs (but not in B cells) was higher in patients with NSCLC alone than in patients with COPD alone. The percentage of APRIL-expressing AECs (but not B cells or AMs) was higher in COPD patients than in SC and NSC (p < 0.05 for all comparisons). The percentage of APRIL-expressing B cells, AMs and AECs cells was similar in NSC and SC. Conclusion The percentage of APRIL-expressing B cells, AMs and AECs is higher in the lungs of patients with both COPD and NSCLC than in patients with COPD or NSCLC alone or control subjects. These findings suggest that APRIL may contribute to the pathogenesis of both COPD and NSCLC, and possibly to the development of NSCLC in patients with established COPD.
Collapse
Affiliation(s)
- Francesca Polverino
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Room 855B, Harvard Institutes of Medicine Building, 77 Avenue Louis Pasteur, Boston, MA 02115 USA.,Lovelace Respiratory Research Institute, Albuquerque, NM USA.,University of Parma, Parma, Italy
| | - Maria Laucho-Contreras
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Room 855B, Harvard Institutes of Medicine Building, 77 Avenue Louis Pasteur, Boston, MA 02115 USA
| | - Joselyn Rojas Quintero
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Room 855B, Harvard Institutes of Medicine Building, 77 Avenue Louis Pasteur, Boston, MA 02115 USA
| | - Miguel Divo
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Room 855B, Harvard Institutes of Medicine Building, 77 Avenue Louis Pasteur, Boston, MA 02115 USA.,Lovelace Respiratory Research Institute, Albuquerque, NM USA
| | - Victor Pinto-Plata
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Room 855B, Harvard Institutes of Medicine Building, 77 Avenue Louis Pasteur, Boston, MA 02115 USA.,Lovelace Respiratory Research Institute, Albuquerque, NM USA
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA USA
| | | | - Bartolome R Celli
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Room 855B, Harvard Institutes of Medicine Building, 77 Avenue Louis Pasteur, Boston, MA 02115 USA.,Lovelace Respiratory Research Institute, Albuquerque, NM USA
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Room 855B, Harvard Institutes of Medicine Building, 77 Avenue Louis Pasteur, Boston, MA 02115 USA.,Lovelace Respiratory Research Institute, Albuquerque, NM USA
| |
Collapse
|
37
|
Significance of BAFF/APRIL Expression and Their Receptors in Pediatric Patients With Acute Lymphoblastic Leukemia. J Pediatr Hematol Oncol 2016; 38:167-72. [PMID: 26950089 DOI: 10.1097/mph.0000000000000549] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
In this study, we investigated the mRNA expression and protein levels of B-cell activating factor (BAFF)/a proliferation-inducing ligand (APRIL) and their receptors in acute lymphoblastic leukemia (ALL) cell lines and pediatric patients with ALL using real-time polymerase chain reaction, enzyme-linked immunosorbent assay, and Western blotting. The location and level of the BAFF/APRIL proteins in ALL cell lines were also detected by immunofluorescence cytochemistry and flow cytometry. Correlations between plasma protein levels of BAFF/APRIL and primary clinical parameters were analyzed. We found that BAFF/APRIL was highly expressed in pediatric ALL patients and ALL cell lines. The BAFF/APRIL proteins were located on the cell membrane, and the proportion of positive cells and mean fluorescence intensity were significantly higher than in the healthy control group (P<0.05). The mRNA expression and protein levels of BAFF/APRIL and their receptors in untreated ALL children were significantly higher than in healthy controls (P<0.05) as well as were significantly reduced in the remission group (P<0.05). The plasma protein levels of BAFF/APRIL were positively correlated with the white blood cell count, lactate dehydrogenase, and serum ferritin. Abnormal levels of BAFF/APRIL in pediatric ALL suggest that BAFF/APRIL are associated with the development and progression of ALL in children and may provide information for the development of BAFF-based and APRIL-based targeted therapies.
Collapse
|
38
|
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease with variable clinical manifestations. While the clearest guidelines for the treatment of SLE exist in the context of lupus nephritis, patients with other lupus manifestations such as neuropsychiatric, hematologic, musculoskeletal, and severe cutaneous lupus frequently require immunosuppression and/or biologic therapy. Conventional immunosuppressive agents such as mycophenolate mofetil, azathioprine, and cyclophosphamide are widely used in the management of SLE with current more rationalized treatment regimens optimizing the use of these agents while minimizing potential toxicity. The advent of biologic therapies has advanced the treatment of SLE particularly in patients with refractory disease. The CD20 monoclonal antibody rituximab and the anti-BLyS agent belimumab are now widely in use in clinical practice. Several other biologic agents are in ongoing clinical trials. While immunosuppressive and biologic agents are the foundation of inflammatory disease control in SLE, the importance of managing comorbidities such as cardiovascular risk factors, bone health, and minimizing susceptibility to infection should not be neglected.
Collapse
Affiliation(s)
- Natasha Jordan
- Department of Rheumatology, Cambridge University Hospitals NHS Foundation Trust, Cambridge
| | - David D'Cruz
- Louise Coote Lupus Unit, Guy's and St Thomas' Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
39
|
Jordan N, Lutalo PM, D'Cruz DP. Progress with the use of monoclonal antibodies for the treatment of systemic lupus erythematosus. Immunotherapy 2016; 7:255-70. [PMID: 25804478 DOI: 10.2217/imt.14.118] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In recent years, significant progress has been made in the use of monoclonal antibodies in the treatment of systemic lupus erythematosus (SLE). Advances in our understanding of the complexity of SLE immunopathogenesis have led to the testing of several biologic agents in clinical trials. Monoclonal therapies currently emerging or under development include B-cell depletion therapies, agents targeting B-cell survival factors, blockade of T-cell co-stimulation and anticytokine therapies. Issues remain, however, regarding clinical trial design and outcome measures in SLE which need to be addressed to optimize translation of these promising therapies into clinical practice.
Collapse
Affiliation(s)
- Natasha Jordan
- Louise Coote Lupus Unit St Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH, UK
| | | | | |
Collapse
|
40
|
Jordan N, D'Cruz D. Key issues in the management of patients with systemic lupus erythematosus: latest developments and clinical implications. Ther Adv Musculoskelet Dis 2015; 7:234-46. [PMID: 26622325 DOI: 10.1177/1759720x15601805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Systemic lupus erythematous (SLE) is a chronic multisystem disease with significant associated morbidity and mortality. A deeper understanding of the pathogenesis of SLE has led to the development of biologic agents, primarily targeting B cells and others inhibiting costimulatory molecules, type I interferons and cytokines such as interleukin-6. Several of these agents have been studied in clinical trials; some have shown promise while others have yielded disappointing results. Economic and regulatory issues continue to hamper the availability of such therapies for SLE patients. With increasing recognition that recurrent flares of disease activity lead to long-term damage accrual, one of the most important recent developments in patient management has been the concept of treat-to-target in SLE while minimizing patient exposure to excessive corticosteroid and other immunosuppressive therapy. This article reviews these key issues in SLE management, outlining recent developments and clinical implications for patients.
Collapse
Affiliation(s)
- Natasha Jordan
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - David D'Cruz
- Louise Coote Lupus Unit, Guys Hospital, London, SE1 9RT, UK
| |
Collapse
|
41
|
Activation of LXR attenuates collagen-induced arthritis via suppressing BLyS production. Clin Immunol 2015; 161:339-47. [PMID: 26431776 DOI: 10.1016/j.clim.2015.09.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 09/16/2015] [Accepted: 09/29/2015] [Indexed: 01/03/2023]
Abstract
B-lymphocyte stimulator (BLyS) plays a critical role in the pathogenesis and progression of rheumatoid arthritis (RA). Liver X receptor (LXR), a nuclear receptor, has an important anti-inflammatory effect. However, it is unclear whether the BLyS expression is regulated by LXR. In this study, we found that treatment with LXR agonist in collagen-induced arthritis (CIA) mice significantly attenuated arthritis progression, and markedly decreased BLyS production in serum and splenocytes as well as the production of serum IFNγ and TGFβ. Activation of LXR in B lymphocytes dramatically suppressed the basal and IFNγ/TGFβ-induced BLyS expression. Moreover, LXR agonist prominently suppressed the binding of NF-κB to BLyS promoter region, and decreased the promoter's transcriptional activity. Additionally, activation of LXR obviously repressed IFNγ-induced STAT1 activation and TGFβ-induced SMAD3 activation. These results indicated that downregulation of BLyS may be a novel mechanism by which LXR ameliorates RA, and LXR/BLyS pathway may serve as a novel target for the treatment of RA.
Collapse
|
42
|
Morais SA, Vilas-Boas A, Isenberg DA. B-cell survival factors in autoimmune rheumatic disorders. Ther Adv Musculoskelet Dis 2015; 7:122-51. [PMID: 26288664 PMCID: PMC4530383 DOI: 10.1177/1759720x15586782] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Autoimmune rheumatic disorders have complex etiopathogenetic mechanisms in which B cells play a central role. The importance of factors stimulating B cells, notably the B-cell activating factor (BAFF) and A proliferation inducing ligand (APRIL) axis is now recognized. BAFF and APRIL are cytokines essential for B-cell proliferation and survival from the immature stages to the development of plasma cells. Their levels are increased in some subsets of patients with autoimmune disorders. Several recent biologic drugs have been developed to block this axis, namely belimumab [already licensed for systemic lupus erythematosus (SLE) treatment], tabalumab, atacicept and blisibimod. Many clinical trials to evaluate the safety and efficacy of these drugs in several autoimmune disorders are ongoing, or have been completed recently. This review updates the information on the use of biologic agents blocking BAFF/APRIL for patients with SLE, rheumatoid arthritis, Sjögren's syndrome and myositis.
Collapse
Affiliation(s)
- Sandra A Morais
- Internal Medicine Department, Hospital Pedro Hispano, Matosinhos, Portugal
| | - Andreia Vilas-Boas
- Internal Medicine Department, Hospital Pedro Hispano, Matosinhos, Portugal
| | - David A Isenberg
- Centre for Rheumatology, University College London, Room 424, 4th Floor Rayne Building, 5 University Street, London WC1E 6JF, UK
| |
Collapse
|
43
|
Relle M, Weinmann-Menke J, Scorletti E, Cavagna L, Schwarting A. Genetics and novel aspects of therapies in systemic lupus erythematosus. Autoimmun Rev 2015; 14:1005-18. [PMID: 26164648 DOI: 10.1016/j.autrev.2015.07.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 07/06/2015] [Indexed: 02/06/2023]
Abstract
Autoimmune diseases, such as rheumatoid arthritis, multiple sclerosis, autoimmune hepatitis and inflammatory bowel disease, have complex pathogeneses and the factors which cause these disorders are not well understood. But all have in common that they arise from a dysfunction of the immune system, interpreting self components as foreign antigens. Systemic lupus erythematosus (SLE) is one of these complex inflammatory disorders that mainly affects women and can lead to inflammation and severe damage of virtually any tissue and organ. Recently, the application of advanced techniques of genome-wide scanning revealed more genetic information about SLE than previously possible. These case-control or family-based studies have provided evidence that SLE susceptibility is based (with a few exceptions) on an individual accumulation of various risk alleles triggered by environmental factors and also help to explain the discrepancies in SLE susceptibility between different populations or ethnicities. Moreover, during the past years new therapies (autologous stem cell transplantation, B cell depletion) and improved conventional treatment options (corticosteroids, traditional and new immune-suppressants like mycophenolate mofetile) changed the perspective in SLE therapeutic approaches. Thus, this article reviews genetic aspects of this autoimmune disease, summarizes clinical aspects of SLE and provides a general overview of conventional and new therapeutic approaches in SLE.
Collapse
Affiliation(s)
- Manfred Relle
- First Department of Medicine, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Julia Weinmann-Menke
- First Department of Medicine, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Eva Scorletti
- Division of Rheumatology, IRCCS Fondazione Policlinico San Matteo, Lombardy, Pavia, Italy
| | - Lorenzo Cavagna
- Division of Rheumatology, IRCCS Fondazione Policlinico San Matteo, Lombardy, Pavia, Italy
| | - Andreas Schwarting
- First Department of Medicine, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany; Acura Centre of Rheumatology Rhineland-Palatinate, Bad Kreuznach, Germany.
| |
Collapse
|
44
|
Schuepbach-Mallepell S, Das D, Willen L, Vigolo M, Tardivel A, Lebon L, Kowalczyk-Quintas C, Nys J, Smulski C, Zheng TS, Maskos K, Lammens A, Jiang X, Hess H, Tan SL, Schneider P. Stoichiometry of Heteromeric BAFF and APRIL Cytokines Dictates Their Receptor Binding and Signaling Properties. J Biol Chem 2015; 290:16330-42. [PMID: 25953898 PMCID: PMC4481231 DOI: 10.1074/jbc.m115.661405] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Indexed: 12/31/2022] Open
Abstract
The closely related TNF family ligands B cell activation factor (BAFF) and a proliferation-inducing ligand (APRIL) serve in the generation and maintenance of mature B-lymphocytes. Both BAFF and APRIL assemble as homotrimers that bind and activate several receptors that they partially share. However, heteromers of BAFF and APRIL that occur in patients with autoimmune diseases are incompletely characterized. The N and C termini of adjacent BAFF or APRIL monomers are spatially close and can be linked to create single-chain homo- or hetero-ligands of defined stoichiometry. Similar to APRIL, heteromers consisting of one BAFF and two APRILs (BAA) bind to the receptors B cell maturation antigen (BCMA), transmembrane activator and CAML interactor (TACI) but not to the BAFF receptor (BAFFR). Heteromers consisting of one APRIL and two BAFF (ABB) bind to TACI and BCMA and weakly to BAFFR in accordance with the analysis of the receptor interaction sites in the crystallographic structure of ABB. Receptor binding correlated with activity in reporter cell line assays specific for BAFFR, TACI, or BCMA. Single-chain BAFF (BBB) and to a lesser extent single-chain ABB, but not APRIL or single-chain BAA, rescued BAFFR-dependent B cell maturation in BAFF-deficient mice. In conclusion, BAFF-APRIL heteromers of different stoichiometries have distinct receptor-binding properties and activities. Based on the observation that heteromers are less active than BAFF, we speculate that their physiological role might be to down-regulate BAFF activity.
Collapse
Affiliation(s)
| | - Dolon Das
- From the Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Laure Willen
- From the Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Michele Vigolo
- From the Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Aubry Tardivel
- From the Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Luc Lebon
- From the Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | | | - Josquin Nys
- From the Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Cristian Smulski
- From the Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | | | - Klaus Maskos
- Proteros Biostructures GmbH, D-82152 Planegg, Germany
| | | | - Xuliang Jiang
- the EMD Serono Research and Development Institute, Billerica, Massachusetts 01821
| | - Henry Hess
- a subsidiary of Merck KGaA, D-64293 Darmstadt, Germany
| | - Seng-Lai Tan
- the EMD Serono Research and Development Institute, Billerica, Massachusetts 01821
| | - Pascal Schneider
- From the Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland,
| |
Collapse
|
45
|
Donahoe M, Valentine VG, Chien N, Gibson KF, Raval JS, Saul M, Xue J, Zhang Y, Duncan SR. Autoantibody-Targeted Treatments for Acute Exacerbations of Idiopathic Pulmonary Fibrosis. PLoS One 2015; 10:e0127771. [PMID: 26083430 PMCID: PMC4470587 DOI: 10.1371/journal.pone.0127771] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 04/18/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Severe acute exacerbations (AE) of idiopathic pulmonary fibrosis (IPF) are medically untreatable and often fatal within days. Recent evidence suggests autoantibodies may be involved in IPF progression. Autoantibody-mediated lung diseases are typically refractory to glucocorticoids and nonspecific medications, but frequently respond to focused autoantibody reduction treatments. We conducted a pilot trial to test the hypothesis that autoantibody-targeted therapies may also benefit AE-IPF patients. METHODS Eleven (11) critically-ill AE-IPF patients with no evidence of conventional autoimmune diseases were treated with therapeutic plasma exchanges (TPE) and rituximab, supplemented in later cases with intravenous immunoglobulin (IVIG). Plasma anti-epithelial (HEp-2) autoantibodies and matrix metalloproteinase-7 (MMP7) were evaluated by indirect immunofluorescence and ELISA, respectively. Outcomes among the trial subjects were compared to those of 20 historical control AE-IPF patients treated with conventional glucocorticoid therapy prior to this experimental trial. RESULTS Nine (9) trial subjects (82%) had improvements of pulmonary gas exchange after treatment, compared to one (5%) historical control. Two of the three trial subjects who relapsed after only five TPE responded again with additional TPE. The three latest subjects who responded to an augmented regimen of nine TPE plus rituximab plus IVIG have had sustained responses without relapses after 96-to-237 days. Anti-HEp-2 autoantibodies were present in trial subjects prior to therapy, and were reduced by TPE among those who responded to treatment. Conversely, plasma MMP7 levels were not systematically affected by therapy nor correlated with clinical responses. One-year survival of trial subjects was 46+15% vs. 0% among historical controls. No serious adverse events were attributable to the experimental medications. CONCLUSION This pilot trial indicates specific treatments that reduce autoantibodies might benefit some severely-ill AE-IPF patients. These findings have potential implications regarding mechanisms of IPF progression, and justify considerations for incremental trials of autoantibody-targeted therapies in AE-IPF patients. TRIAL REGISTRATION ClinicalTrials.gov NCT01266317.
Collapse
Affiliation(s)
- Michael Donahoe
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213, United States of America
| | - Vincent G. Valentine
- Department of Medicine, University of Texas Medical Branch, Galveston, Texas, 77555, United States of America
| | - Nydia Chien
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213, United States of America
| | - Kevin F. Gibson
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213, United States of America
| | - Jay S. Raval
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, 27599, United States of America
| | - Melissa Saul
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213, United States of America
| | - Jianmin Xue
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213, United States of America
| | - Yingze Zhang
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213, United States of America
| | - Steven R. Duncan
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213, United States of America
- * E-mail:
| |
Collapse
|
46
|
Effect of TACI signaling on humoral immunity and autoimmune diseases. J Immunol Res 2015; 2015:247426. [PMID: 25866827 PMCID: PMC4381970 DOI: 10.1155/2015/247426] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/04/2015] [Indexed: 02/02/2023] Open
Abstract
Transmembrane activator and calcium-modulating cyclophilin ligand interactor (TACI) is one of the receptors of B cell activating factor of the tumor necrosis factor family (BAFF) and a proliferation-inducing ligand (APRIL). TACI is a regulator in the immune responses. TACI inhibits B cell expansion and promotes the differentiation and survival of plasma cells. The mechanisms underlying these effects probably involve changed expressions of some crucial molecules, such as B lymphocyte induced maturation protein-1 (Blimp-1) and inducible T-cell costimulator ligand (ICOSL) in B cells and/or plasma cells. However, abnormal TACI signaling may relate to autoimmune disorders. Common variable immune deficiency (CVID) patients with heterozygous mutations in TACI alleles increase susceptibility to autoimmune diseases. Taci−/− mice and BAFF transgenic mice both develop signs of human SLE. These findings that indicate inappropriate levels of TACI signaling may disrupt immune system balance, thereby promoting the development of autoimmune diseases. In this review, we summarize the basic characteristics of the TACI ligands BAFF and APRIL, and detail the research findings on the role of TACI in humoral immunity. We also discuss the possible mechanisms underlying the susceptibility of CVID patients with TACI mutations to autoimmune diseases and the role of TACI in the pathogenesis of SLE.
Collapse
|
47
|
Han L, Zhang W, Song F, Guo Y, Guo K, Zhou W. Soluble a‑proliferation‑inducing ligand (sAPRIL), a novel serum biomarker predicting the recurrence and metastasis of pancreatic adenocarcinoma after surgery. Mol Med Rep 2014; 10:1978-84. [PMID: 25110120 DOI: 10.3892/mmr.2014.2443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 05/19/2014] [Indexed: 11/06/2022] Open
Abstract
Pancreatic adenocarcinoma (PA) is a leading cause of adult cancer mortality, and surgery is still the best available treatment strategy. However, PA can recur at any time and has limited prognosis. It is therefore necessary to explore novel serum biomarkers of PA to allow the early diagnosis of PA. Soluble a-proliferation-inducing ligand (sAPRIL), a promising inducer of the epithelial-mesenchymal transition (EMT), is often found overexpressed in a variety of autoimmune diseases. To determine whether serum sAPRIL can constitute a PA biomarker, the protein level of sAPRIL was examined by immunohistochemistry and western blot, and the mRNA level was quantified by RT-qPCR. The PA cell line PanC-1 was transfected with vectors bearing the sAPRIL gene and sAPRIL short hairpin RNA (shRNA) oligos. Increased expression of serum sAPRIL was observed in patients with PA recurrence or metastasis after five-year surgery compared to subjects without PA recurrence or metastasis. The growth rate of PanC-1 cells transfected with the sAPRIL expression vector was increased by 23% (P<0.01, vs. control group), and was reduced by 17% (P<0.01, vs. control group) in the sAPRIL shRNA-silenced cell line. Thus, sAPRIL is highly expressed in PA, and serum levels of sAPRIL can serve as a useful indicator for the recurrence or metastasis of PA after surgery. Additional validation studies on the use of serum sAPRIL as a diagnostic marker in PA are however needed.
Collapse
Affiliation(s)
- Lei Han
- Department of Hepatobiliary and Pancreas Surgery, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, P.R. China
| | - Wei Zhang
- Department of Hepatobiliary and Pancreas Surgery, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, P.R. China
| | - Fulin Song
- Department of Pathology, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, P.R. China
| | - Yang Guo
- Department of Hepatobiliary and Pancreas Surgery, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, P.R. China
| | - Kejian Guo
- Department of General Surgery, College of Clinical Medical Sciences, China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Wenping Zhou
- Department of Hepatobiliary and Pancreas Surgery, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
48
|
Eilertsen GØ, Nossent JC. APRIL levels strongly correlate with IL-17 in systemic lupus erythematosus. Lupus 2014; 23:1383-91. [DOI: 10.1177/0961203314543914] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Introduction Activated self-reactive B cells play an important part in systemic lupus erythematosus (SLE). A proliferation-inducing ligand (APRIL) and B cell-activating factor (BAFF) are B-cell specific stimulators, but activate B cells through different receptors. We investigated the reciprocal association between serum APRIL (s-APRIL), serum BAFF (s-BAFF) and immunological and clinical findings in SLE patients. Methods A cross-sectional case-control study was performed in 100 SLE patients (87% female, age 49 years, disease duration 12 years). APRIL and BAFF levels were measured by sandwich ELISA, compared with healthy controls and correlated with autoantibody, cytokine (IL-6 and IL-17) and clinical findings through nonparametric and multivariate regression analyses. Results Both median s-APRIL (478 vs. 0 pg/ml, p = 0.01) and s-BAFF (1720 vs. 0.9 pg/ml, p < 0.001) were higher in SLE patients than controls. Increased s-BAFF was observed in 86% of patients, while s-APRIL was increased only in 17% ( p < 0.01). S-APRIL correlated with s-BAFF in controls ( p = 0.04), but not in SLE ( p = 0.8). Increased s-APRIL was strongly and independently associated with IL-17 activation ( p < 0.001), while increased s-BAFF levels were associated with anti-nucleosome antibody presence ( p = 0.001). Disease activity and organ damage were associated with s-BAFF but not s-APRIL. Conclusions While both s-BAFF and s-APRIL levels are elevated in SLE patients, they reflect different immunologic and clinical pathways. The strong association between s-APRIL and IL-17 activation supports a role for Th17 helper cells in B cell activation in SLE.
Collapse
Affiliation(s)
- GØ Eilertsen
- Bone and Joint Research Group, Department of Clinical Medicine, Faculty of Health Science, University of Tromsø, Norway
| | - J C Nossent
- Bone and Joint Research Group, Department of Clinical Medicine, Faculty of Health Science, University of Tromsø, Norway
- Rheumatology Section, Division of Medicine, Royal Darwin Hospital, Darwin, NT, Australia
| |
Collapse
|
49
|
Munroe ME, Vista ES, Guthridge JM, Thompson LF, Merrill JT, James JA. Proinflammatory adaptive cytokine and shed tumor necrosis factor receptor levels are elevated preceding systemic lupus erythematosus disease flare. Arthritis Rheumatol 2014; 66:1888-99. [PMID: 24578190 PMCID: PMC4128244 DOI: 10.1002/art.38573] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 02/20/2014] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a multifaceted disease characterized by immune dysregulation and unpredictable disease activity. This study sought to evaluate the changes in plasma concentrations of soluble mediators that precede clinically defined disease flares. METHODS Fifty-two different soluble mediators, including cytokines, chemokines, and soluble receptors, were examined using validated multiplex bead-based or enzyme-linked immunosorbent assays in plasma from 28 European American patients with SLE who developed disease flare 6 or 12 weeks after a baseline assessment (preflare), 28 matched SLE patients without impending flare (nonflare), and 28 matched healthy controls. In a subset of 13 SLE patients, mediators within samples obtained preceding disease flare were compared with those within samples from the same individual obtained during a clinically stable period without flare. RESULTS Compared to SLE patients with clinically stable disease, SLE patients with impending flare had significant alterations (P ≤ 0.01) in the levels of 27 soluble mediators at baseline; specifically, the levels of proinflammatory mediators, including Th1-, Th2-, and Th17-type cytokines, were significantly higher several weeks before clinical flare. Baseline levels of regulatory cytokines, including interleukin-10 and transforming growth factor β, were higher in nonflare SLE patients, whereas baseline levels of soluble tumor necrosis factor receptor type I (TNFRI), TNFRII, Fas, FasL, and CD40L were significantly higher (P ≤ 0.002) in preflare SLE patients. The normalized and weighted combined soluble mediator score was significantly higher (P ≤ 0.0002) in preflare samples from SLE patients compared to samples from the same patients obtained during periods of stable disease. CONCLUSION The levels of proinflammatory adaptive cytokines and shed TNF receptors are elevated prior to disease flare, while the levels of regulatory mediators are elevated during periods of stable disease. Alterations in the balance between inflammatory and regulatory mediators may help identify patients at risk of disease flare and help decipher the pathogenic mechanisms of SLE.
Collapse
MESH Headings
- Adaptive Immunity/immunology
- Adult
- Biomarkers/blood
- CD40 Ligand/blood
- CD40 Ligand/immunology
- Cytokines/blood
- Cytokines/immunology
- Fas Ligand Protein/blood
- Fas Ligand Protein/immunology
- Female
- Humans
- Inflammation Mediators/blood
- Inflammation Mediators/immunology
- Lupus Erythematosus, Systemic/epidemiology
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Middle Aged
- Receptors, Tumor Necrosis Factor, Type I/blood
- Receptors, Tumor Necrosis Factor, Type I/immunology
- Receptors, Tumor Necrosis Factor, Type II/blood
- Receptors, Tumor Necrosis Factor, Type II/immunology
- Risk Factors
- Severity of Illness Index
- fas Receptor/blood
- fas Receptor/immunology
Collapse
Affiliation(s)
- Melissa E. Munroe
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Evan S. Vista
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Division of Rheumatology, University of Santo Tomas Hospital, Manila, PH
| | - Joel M. Guthridge
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Linda F. Thompson
- Immunobiology and Cancer, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Joan T. Merrill
- Clinical Pharmacology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Judith A. James
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Departments of Medicine, Pathology, and Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
50
|
Specific forms of BAFF favor BAFF receptor-mediated epithelial cell survival. J Autoimmun 2014; 51:30-7. [DOI: 10.1016/j.jaut.2014.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/05/2014] [Indexed: 01/23/2023]
|