1
|
Jinshan Z, Yong Q, Fangqi C, Juanmei C, Min L, Changzheng H. The role of TNF-α as a potential marker for acute cutaneous lupus erythematosus in patients with systemic lupus erythematosus. J Dermatol 2024; 51:1481-1491. [PMID: 38963308 DOI: 10.1111/1346-8138.17355] [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: 01/28/2024] [Revised: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 07/05/2024]
Abstract
Acute cutaneous lupus erythematosus (ACLE) is closely associated with systemic symptoms in systemic lupus erythematosus (SLE). This study aimed to identify potential biomarkers for ACLE and explore their association with SLE to enable early prediction of ACLE and identify potential treatment targets for the future. In total, 185 SLE-diagnosed patients were enrolled and categorized into two groups: those with ACLE and those without cutaneous involvement. After conducting logistic regression analysis of the differentiating factors, we concluded that tumor necrosis factor-alpha (TNF-α) is an independent risk factor for ACLE. Analysis of the receiver operating characteristic revealed an area under the curve of 0.716 for TNF-α. Additionally, both TNF-α and ACLE are positively correlated with disease activity. TNF-α shows promise as a biomarker for ACLE, and in SLE patients, ACLE may serve as a clear indicator of moderate-to-severe disease activity.
Collapse
Affiliation(s)
- Zhan Jinshan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qu Yong
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chen Fangqi
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Cao Juanmei
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Department of Dermatology, The First Affiliated Hospital of Shihezi University, Shihezi University, Shihezi, Xinjiang, China
| | - Li Min
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Huang Changzheng
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Pandey SP, Bhaskar R, Han SS, Narayanan KB. Autoimmune Responses and Therapeutic Interventions for Systemic Lupus Erythematosus: A Comprehensive Review. Endocr Metab Immune Disord Drug Targets 2024; 24:499-518. [PMID: 37718519 DOI: 10.2174/1871530323666230915112642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 06/05/2023] [Accepted: 07/22/2023] [Indexed: 09/19/2023]
Abstract
Systemic Lupus Erythematosus (SLE) or Lupus is a multifactorial autoimmune disease of multiorgan malfunctioning of extremely heterogeneous and unclear etiology that affects multiple organs and physiological systems. Some racial groups and women of childbearing age are more susceptible to SLE pathogenesis. Impressive progress has been made towards a better understanding of different immune components contributing to SLE pathogenesis. Recent investigations have uncovered the detailed mechanisms of inflammatory responses and organ damage. Various environmental factors, pathogens, and toxicants, including ultraviolet light, drugs, viral pathogens, gut microbiome metabolites, and sex hormones trigger the onset of SLE pathogenesis in genetically susceptible individuals and result in the disruption of immune homeostasis of cytokines, macrophages, T cells, and B cells. Diagnosis and clinical investigations of SLE remain challenging due to its clinical heterogeneity and hitherto only a few approved antimalarials, glucocorticoids, immunosuppressants, and some nonsteroidal anti-inflammatory drugs (NSAIDs) are available for treatment. However, the adverse effects of renal and neuropsychiatric lupus and late diagnosis make therapy challenging. Additionally, SLE is also linked to an increased risk of cardiovascular diseases due to inflammatory responses and the risk of infection from immunosuppressive treatment. Due to the diversity of symptoms and treatment-resistant diseases, SLE management remains a challenging issue. Nevertheless, the use of next-generation therapeutics with stem cell and gene therapy may bring better outcomes to SLE treatment in the future. This review highlights the autoimmune responses as well as potential therapeutic interventions for SLE particularly focusing on the recent therapeutic advancements and challenges.
Collapse
Affiliation(s)
- Surya Prakash Pandey
- Aarogya Institute of Healthcare and Research, Jaipur, Rajasthan, 302033, India
- Department of Zoology, School of Science, IFTM University, Moradabad, Uttar Pradesh, 244102, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| | - Kannan Badri Narayanan
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| |
Collapse
|
3
|
Mukherjee I, Singh S, Karmakar A, Kashyap N, Mridha AR, Sharma JB, Luthra K, Sharma RS, Biswas S, Dhar R, Karmakar S. New immune horizons in therapeutics and diagnostic approaches to Preeclampsia. Am J Reprod Immunol 2023; 89:e13670. [PMID: 36565013 DOI: 10.1111/aji.13670] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 11/02/2022] [Accepted: 12/10/2022] [Indexed: 12/25/2022] Open
Abstract
Hypertensive disorders of pregnancy (HDP) are one of the commonest maladies, affecting 5%-10% of pregnancies worldwide. The American College of Obstetricians and Gynecologists (ACOG) identifies four categories of HDP, namely gestational hypertension (GH), Preeclampsia (PE), chronic hypertension (CH), and CH with superimposed PE. PE is a multisystem, heterogeneous disorder that encompasses 2%-8% of all pregnancy-related complications, contributing to about 9% to 26% of maternal deaths in low-income countries and 16% in high-income countries. These translate to 50 000 maternal deaths and over 500 000 fetal deaths worldwide, therefore demanding high priority in understanding clinical presentation, screening, diagnostic criteria, and effective management. PE is accompanied by uteroplacental insufficiency leading to vascular and metabolic changes, vasoconstriction, and end-organ ischemia. PE is diagnosed after 20 weeks of pregnancy in women who were previously normotensive or hypertensive. Besides shallow trophoblast invasion and inadequate remodeling of uterine arteries, dysregulation of the nonimmune system has been the focal point in PE. This results from aberrant immune system activation and imbalanced differentiation of T cells. Further, a failure of tolerance toward the semi-allogenic fetus results due to altered distribution of Tregs such as CD4+FoxP3+ or CD4+CD25+CD127(low) FoxP3+ cells, thereby creating a cytotoxic environment by suboptimal production of immunosuppressive cytokines like IL-10, IL-4, and IL-13. Also, intracellular production of complement protein C5a may result in decreased FoxP3+ regulatory T cells. With immune system dysfunction as a major driver in PE pathogenesis, it is logical that therapeutic targeting of components of the immune system with pharmacologic agents like anti-inflammatory and immune-modulating molecules are either being used or under clinical trial. Cholesterol synthesis inhibitors like Pravastatin may improve placental perfusion in PE, while Eculizumab (monoclonal antibody inhibiting C5) and small molecular inhibitor of C5a, Zilucoplan are under investigation. Monoclonal antibody against IL-17(Secukinumab) has been proposed to alter the Th imbalance in PE. Autologous Treg therapy and immune checkpoint inhibitors like anti-CTLA-4 are emerging as new candidates in immune horizons for PE management in the future.
Collapse
Affiliation(s)
- Indrani Mukherjee
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.,Amity Institute of Biotechnology (AIB), Amity University, Noida, India
| | - Sunil Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Abhibrato Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Neha Kashyap
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Asit Ranjan Mridha
- Department of Obstetrics & Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Jai Bhagwan Sharma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Radhey Shyam Sharma
- Ex-Head and Scientist G, Indian Council of Medical Research, New Delhi, India
| | - Subhrajit Biswas
- Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University, Noida, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
4
|
Ghorbaninezhad F, Leone P, Alemohammad H, Najafzadeh B, Nourbakhsh NS, Prete M, Malerba E, Saeedi H, Tabrizi NJ, Racanelli V, Baradaran B. Tumor necrosis factor‑α in systemic lupus erythematosus: Structure, function and therapeutic implications (Review). Int J Mol Med 2022; 49:43. [PMID: 35137914 DOI: 10.3892/ijmm.2022.5098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/05/2022] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor‑α (TNF‑α) is a pleiotropic pro‑inflammatory cytokine that contributes to the pathophysiology of several autoimmune diseases, such as multiple sclerosis, inflammatory bowel disease, rheumatoid arthritis, psoriatic arthritis and systemic lupus erythematosus (SLE). The specific role of TNF‑α in autoimmunity is not yet fully understood however, partially, in a complex disease such as SLE. Through the engagement of the TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2), both the two variants, soluble and transmembrane TNF‑α, can exert multiple biological effects according to different settings. They can either function as immune regulators, impacting B‑, T‑ and dendritic cell activity, modulating the autoimmune response, or as pro‑inflammatory mediators, regulating the induction and maintenance of inflammatory processes in SLE. The present study reviews the dual role of TNF‑α, focusing on the different effects that TNF‑α may have on the pathogenesis of SLE. In addition, the efficacy and safety of anti‑TNF‑α therapies in preclinical and clinical trials SLE are discussed.
Collapse
Affiliation(s)
- Farid Ghorbaninezhad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan 5165665811, Iran
| | - Patrizia Leone
- Department of Biomedical Sciences and Human Oncology, 'Aldo Moro' University of Bari Medical School, I‑70124 Bari, Italy
| | - Hajar Alemohammad
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, East Azerbaijan 5166616471, Iran
| | - Basira Najafzadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, East Azerbaijan 5166616471, Iran
| | - Niloufar Sadat Nourbakhsh
- Department of Genetics, Faculty of Basic Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Fars 7319846451, Iran
| | - Marcella Prete
- Department of Biomedical Sciences and Human Oncology, 'Aldo Moro' University of Bari Medical School, I‑70124 Bari, Italy
| | - Eleonora Malerba
- Department of Biomedical Sciences and Human Oncology, 'Aldo Moro' University of Bari Medical School, I‑70124 Bari, Italy
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan 5165665811, Iran
| | - Neda Jalili Tabrizi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan 5165665811, Iran
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, 'Aldo Moro' University of Bari Medical School, I‑70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan 5165665811, Iran
| |
Collapse
|
5
|
Said JT, Elman SA, Merola JF. Evaluating safety and compatibility of anti-tumor necrosis factor therapy in patients with connective tissue disorders. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:430. [PMID: 33842651 PMCID: PMC8033307 DOI: 10.21037/atm-20-5552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Inhibition of the proinflammatory cytokine tumor necrosis factor alpha (TNFα) has been utilized as a treatment strategy for a variety of immune-mediated inflammatory disorders (IMID), including rheumatoid arthritis, Crohn’s disease and psoriasis. A wide array of biologic therapies targeting the TNFα molecule, including etanercept, infliximab, certolizumab, golimumab and adalimumab, are routinely used in the care of patients with these conditions. In addition to their therapeutic potential, anti-TNFα agents commonly induce the formation of autoantibodies such as anti-nuclear antibodies and anti-double stranded DNA antibodies; however, the vast majority of these are of IgM isotype and of unclear clinical significance, uncommonly leading to drug-induced autoimmune disease. For these reasons, TNFα inhibition has been a controversial strategy in the treatment of primary connective tissue disorders (CTDs). However, as new therapeutics continue to be developed for the management of CTDs, the potential utility for anti-TNFα agents has become of great interest, demonstrated in several recent case series and small open-label trials. We review the safety and compatibility of anti-TNFα therapy in the management of systemic lupus erythematosus (SLE) and cutaneous lupus erythematosus (CLE), two well-studied example CTDs, as well as summarize the risks of autoantibody generation, infection, malignancy, and iatrogenic lupus flares as side effects of blocking TNFα in patients with these conditions.
Collapse
Affiliation(s)
- Jordan T Said
- Department of Dermatology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Scott A Elman
- Department of Dermatology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Joseph F Merola
- Department of Dermatology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Lorenzo-Vizcaya A, Isenberg DA. The use of anti-TNF-alpha therapies for patients with systemic lupus erythematosus. Where are we now? Expert Opin Biol Ther 2021; 21:639-647. [PMID: 33216641 DOI: 10.1080/14712598.2021.1853096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is an autoimmune rheumatic disease characterized by multiple pathologies in which sustained inflammatory activity leads to progressive tissue destruction and organ damage. One of the main proinflammatory cytokines playing a key role in autoimmune diseases such as rheumatoid arthritis (RA) or SLE, is tumor necrosis factor (TNF) alpha. AREAS COVERED The introduction of TNF-alpha inhibitors revolutionized the treatment of RA and other conditions including psoriatic arthritis and ankylosing spodylitis. We review here the efficacy and safety of TNF-alpha blockers in SLE focussing on why it has not been more widely used since TNF-alpha was reported to be increased in SLE patients and to correlate with disease activity. EXPERT OPINION We summarize the reported SLE cases that have received TNF-alpha blockers and the main results to date. We reflect on whether there is a case to reconsider the use of TNF-alpha blockade in SLE.
Collapse
Affiliation(s)
- Ana Lorenzo-Vizcaya
- Department of Internal Medicine, Hospital Universitario De Ourense. Ourense, Spain
| | - David A Isenberg
- Department of Rheumatology, Division of Medicine, University College London. London, UK
| |
Collapse
|
7
|
Mejia-Vilet JM, Parikh SV, Song H, Fadda P, Shapiro JP, Ayoub I, Yu L, Zhang J, Uribe-Uribe N, Rovin BH. Immune gene expression in kidney biopsies of lupus nephritis patients at diagnosis and at renal flare. Nephrol Dial Transplant 2020; 34:1197-1206. [PMID: 29800348 DOI: 10.1093/ndt/gfy125] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Up to 50% of lupus nephritis (LN) patients experience renal flares after their initial episode of LN. These flares contribute to poor renal outcomes. We postulated that intrarenal immune gene expression is different in flares compared with de novo LN, and conducted these studies to test this hypothesis. METHODS Glomerular and tubulointerstitial immune gene expression was evaluated in 14 patients who had a kidney biopsy to diagnose LN and another biopsy at their first LN flare. Ten healthy living kidney donors were included as controls. RNA was extracted from laser microdissected formalin-fixed paraffin-embedded kidney biopsies. Gene expression was analyzed using the Nanostring nCounter® platform and validated by quantitative real-time polymerase chain reaction. Differentially expressed genes were analyzed by the Ingenuity Pathway Analysis and Panther Gene Ontology tools. RESULTS Over 110 genes were differentially expressed between LN and healthy control kidney biopsies. Although there was considerable molecular heterogeneity between LN biopsies at diagnosis and flare, for about half the LN patients gene expression from the first LN biopsy clustered with the repeated LN biopsy. However, in all patients, a set of eight interferon alpha-controlled genes had a significantly higher expression in the diagnostic biopsy compared with the flare biopsy. In contrast, nine tumor necrosis factor alpha-controlled genes had higher expression in flare biopsies. CONCLUSIONS There is significant heterogeneity in immune-gene expression of kidney tissue from LN patients. There are limited but important differences in gene expression between LN flares, which may influence treatment decisions.
Collapse
Affiliation(s)
- Juan M Mejia-Vilet
- Division of Nephrology, Department of Internal Medicine, Davis Heart and Lung Research Institute, Ohio State University Medical Center, Columbus, OH, USA.,Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Samir V Parikh
- Division of Nephrology, Department of Internal Medicine, Davis Heart and Lung Research Institute, Ohio State University Medical Center, Columbus, OH, USA
| | - Huijuan Song
- Division of Nephrology, Department of Internal Medicine, Davis Heart and Lung Research Institute, Ohio State University Medical Center, Columbus, OH, USA
| | - Paolo Fadda
- Genomics Shared Resource (GSR)-Comprehensive Cancer Center (CCC)
| | - John P Shapiro
- Division of Nephrology, Department of Internal Medicine, Davis Heart and Lung Research Institute, Ohio State University Medical Center, Columbus, OH, USA
| | - Isabelle Ayoub
- Division of Nephrology, Department of Internal Medicine, Davis Heart and Lung Research Institute, Ohio State University Medical Center, Columbus, OH, USA
| | - Lianbo Yu
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jianying Zhang
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Norma Uribe-Uribe
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Brad H Rovin
- Division of Nephrology, Department of Internal Medicine, Davis Heart and Lung Research Institute, Ohio State University Medical Center, Columbus, OH, USA
| |
Collapse
|
8
|
Aringer M. Inflammatory markers in systemic lupus erythematosus. J Autoimmun 2019; 110:102374. [PMID: 31812331 DOI: 10.1016/j.jaut.2019.102374] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 11/20/2019] [Indexed: 01/04/2023]
Abstract
While systemic lupus erythematosus (SLE) is an autoantibody and immune complex disease by nature, most of its organ manifestations are in fact inflammatory. SLE activity scores thus heavily rely on assessing inflammation in the various organs. This focus on clinical items demonstrates that routine laboratory markers of inflammation are still limited in their impact. The erythrocyte sedimentation rate (ESR) is used, but represents a rather crude overall measure. Anemia and diminished serum albumin play a role in estimating inflammatory activity, but both are reflecting more than one mechanism, and the association with inflammation is complex. C-reactive protein (CRP) is a better marker for infections than for SLE activity, where there is only a limited association, and procalcitonin (PCT) is also mainly used for detecting severe bacterial infection. Of the cytokines directly induced by immune complexes, type I interferons, interleukin-18 (IL-18) and tumor necrosis factor (TNF) are correlated with inflammatory disease activity. Still, precise and timely measurement is an issue, which is why they are not currently used for routine purposes. While somewhat more robust in the assays, IL-18 binding protein (IL-18BP) and soluble TNF-receptor 2 (TNF-R2), which are related to the respective cytokines, have not yet made it into clinical routine. The same is true for several chemokines that are increased with activity and relatively easy to measure, but still experimental parameters. In the urine, proteinuria leads and is essential for assessing kidney involvement, but may also result from damage. Similar to the situation in serum and plasma, several cytokines and chemokines perform reasonably well in scientific studies, but are not routine parameters. Cellular elements in the urine are more difficult to assess in the routine laboratory, where sufficient routine is not always available. Therefore, the analysis of urinary T cells may have potential for better monitoring renal inflammation.
Collapse
Affiliation(s)
- Martin Aringer
- University Medical Center and Faculty of Medicine Carl Gustav Carus at the TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
| |
Collapse
|
9
|
Felten R, Dervovic E, Chasset F, Gottenberg JE, Sibilia J, Scher F, Arnaud L. The 2018 pipeline of targeted therapies under clinical development for Systemic Lupus Erythematosus: a systematic review of trials. Autoimmun Rev 2018; 17:781-790. [PMID: 29885544 DOI: 10.1016/j.autrev.2018.02.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 02/03/2018] [Indexed: 12/22/2022]
Abstract
Currently, Systemic Lupus Erythematosus (SLE) therapies range from antimalarials to glucocorticoids, in addition to immunosupressive agents or biologics such as rituximab or belimumab, when needed. Several unmet needs remain in the treatment SLE and more targeted drugs with improved safety profiles are expected. Based on recent advances in the understanding of the complex pathogenesis of SLE, several targeted treatments are currently assessed in clinical trials. In this study, we performed a systematic review of all targeted therapies under clinical development in SLE in 17 online registries of clinical trials. The search yielded a total of 1140 trials, from which we identified 74 targeted therapies for SLE. Those treatments target inflammatory cytokines, chemokines, or their receptors (n = 17), B cells or plasma cells (n = 17), intracellular signalling pathways (n = 10), T/B cells costimulation molecules (n = 8), interferons (n = 7), plasmacytoid dendritic cells (pDC) (n = 3), as well as various other targets (n = 12). Not all these candidate drugs will reach phase III, but the broad spectrum of drugs being investigated may satisfy the urgent need for improved lupus medications. The identification of biomarkers that would allow adequate prediction of response-to-therapy remains high, but when solved will allow a more rationale selection of the optimal pharmacological agent at the patient level.
Collapse
Affiliation(s)
- Renaud Felten
- Service de Rhumatologie, Hôpitaux Universitaires de Strasbourg, RESO, Laboratoire d'Immunopathologie et de Chimie Thérapeutique, Institut de Biologie Moléculaire et Cellulaire (IBMC), CNRS UPR3572, France
| | - Elida Dervovic
- Service de Pharmacie-Stérilisation, Hôpitaux Universitaires de Strasbourg, France
| | - François Chasset
- Sorbonne Université, Faculté de Médecine Sorbonne Université, AP-HP, Service de Dermatologie et Allergologie, Hôpital Tenon, F-75020 Paris, France
| | - Jacques-Eric Gottenberg
- Service de Rhumatologie, Hôpitaux Universitaires de Strasbourg, RESO, Laboratoire d'Immunopathologie et de Chimie Thérapeutique, Institut de Biologie Moléculaire et Cellulaire (IBMC), CNRS UPR3572, France
| | - Jean Sibilia
- Service de Rhumatologie, Hôpitaux Universitaires de Strasbourg, INSERM UMR_S1109, RESO, Université de Strasbourg, F-67000 Strasbourg, France
| | - Florence Scher
- Service de Pharmacie-Stérilisation, Hôpitaux Universitaires de Strasbourg, France
| | - Laurent Arnaud
- Service de Rhumatologie, Hôpitaux Universitaires de Strasbourg, INSERM UMR_S1109, RESO, Université de Strasbourg, F-67000 Strasbourg, France.
| |
Collapse
|
10
|
A New Venue of TNF Targeting. Int J Mol Sci 2018; 19:ijms19051442. [PMID: 29751683 PMCID: PMC5983675 DOI: 10.3390/ijms19051442] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 04/25/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022] Open
Abstract
The first Food and Drug Administration-(FDA)-approved drugs were small, chemically-manufactured and highly active molecules with possible off-target effects, followed by protein-based medicines such as antibodies. Conventional antibodies bind a specific protein and are becoming increasingly important in the therapeutic landscape. A very prominent class of biologicals are the anti-tumor necrosis factor (TNF) drugs that are applied in several inflammatory diseases that are characterized by dysregulated TNF levels. Marketing of TNF inhibitors revolutionized the treatment of diseases such as Crohn’s disease. However, these inhibitors also have undesired effects, some of them directly associated with the inherent nature of this drug class, whereas others are linked with their mechanism of action, being pan-TNF inhibition. The effects of TNF can diverge at the level of TNF format or receptor, and we discuss the consequences of this in sepsis, autoimmunity and neurodegeneration. Recently, researchers tried to design drugs with reduced side effects. These include molecules with more specificity targeting one specific TNF format or receptor, or that neutralize TNF in specific cells. Alternatively, TNF-directed biologicals without the typical antibody structure are manufactured. Here, we review the complications related to the use of conventional TNF inhibitors, together with the anti-TNF alternatives and the benefits of selective approaches in different diseases.
Collapse
|
11
|
Disruption of CXCR3 function impedes the development of Sjögren's syndrome-like xerostomia in non-obese diabetic mice. J Transl Med 2018; 98:620-628. [PMID: 29348563 PMCID: PMC7650019 DOI: 10.1038/s41374-017-0013-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 11/07/2017] [Accepted: 12/05/2017] [Indexed: 11/09/2022] Open
Abstract
The chemokine receptor CXCR3 plays an important role in T cell recruitment in various immune responses and autoimmune diseases. Expression of CXCR3 ligands, including CXCL9, CXCL10, and CXCL11, is elevated in the salivary glands of patients with Sjögren's syndrome (SS). To elucidate whether interaction between CXCR3 and its ligands is required for the development of SS, we administrated an anti-CXCR3 blocking antibody (CXCR3-173) to the non-obese diabetic (NOD) mice, a well-defined model of SS, during the stage prior to disease onset. Treatment with this anti-CXCR3 antibody significantly improved salivary secretion, indicating a remission of SS clinical manifestation. Anti-CXCR3 treatment did not affect the gross leukocyte infiltration of the submandibular glands (SMGs) as assessed by hematoxylin and eosin staining. However, flow cytometric analysis showed that anti-CXCR3 treatment markedly reduced the percentage of CXCR3+CD8 T and CXCR3+CD44+CD8 T cells, without affecting that of CXCR3+CD4 T and CXCR3+CD44+CD4 T cells in the SMGs and submandibular lymph nodes, suggesting a preferential effect of this anti-CXCR3 treatment on CXCR3-expressing effector CD8 T cells. Meanwhile, SMG expression of inflammatory factor TNF-α was markedly diminished by anti-CXCR3 treatment. In accordance, anti-CXCR3 significantly enhanced SMG expression of tight junction protein claudin-1 and water channel protein aquaporin 5, two molecules that are crucial for normal salivary secretion and can be down-regulated by TNF-α. Taken together, these findings demonstrated that the interaction between the endogenous CXCR3 and its ligands plays a pro-inflammatory and pathogenic role in the development of SS-like xerostomia in the NOD mouse model.
Collapse
|
12
|
Danion F, Sparsa L, Arnaud L, Alsaleh G, Lefebvre F, Gies V, Martin T, Lukas C, Durckel J, Ardizzone M, Javier RM, Kleinmann JF, Moreau P, Blaison G, Goetz J, Chatelus E, Gottenberg JE, Sibilia J, Sordet C. Long-term efficacy and safety of antitumour necrosis factor alpha treatment in rhupus: an open-label study of 15 patients. RMD Open 2017; 3:e000555. [PMID: 29435362 PMCID: PMC5761296 DOI: 10.1136/rmdopen-2017-000555] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 10/04/2017] [Accepted: 10/21/2017] [Indexed: 11/03/2022] Open
Abstract
Background The efficacy of antitumour necrosis factor alpha (anti-TNF-α) treatment is well recognised in rheumatoid arthritis (RA) but remains controversial in systemic lupus erythematosus (SLE). Therefore, the role of anti-TNF-α treatment in 'Rhupus', a disease sharing features of RA and SLE, is still debated. Objective To evaluate the efficacy and tolerance of anti-TNF-α in patients with rhupus. Methods Fifteen patients with rhupus with Disease Activity Score 28 (DAS 28) >3.2 despite conventional disease-modifying anti-rheumatic drugs were included in an open-label study. Patients were monitored at months (M) 3, 6, 12, 24 and 60 with SLE Disease Activity Index (SLEDAI) and DAS 28. Statistical analyses were performed using Bayesian methods and Prob >97.5% was considered significant. Results Twelve patients were treated with etanercept for a median duration of 62.5 (range: 6-112) months and three patients by adalimumab during 36.0 (range: 4-52) months. At baseline, median DAS 28 and SLEDAI were 5.94 (4.83-8.09) and 6 (4-8), respectively. DAS 28 and SLEDAI decreased significantly after 3 months, respectively, to 3.70 (1.80-6.42) and 4 (0-6) (Prob >99.9%, for both). These changes persisted at M6, M12, M24 and M60 (Prob >99.9%, for all). Median prednisone dose decreased significantly from 15 (5-35) mg/day to 5 (0-20) mg/day after 6 months and over the follow-up (Prob >99.9%, for all). Tolerance was acceptable, with a severe infection rate of 3.0 per 100 patient-years. Conclusion This pilot study suggests that anti-TNF-α is effective in patients with rhupus with refractive arthritis and has an acceptable safety profile.
Collapse
Affiliation(s)
- François Danion
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Laetitia Sparsa
- Department of Rheumatology, Centre de compétence maladies rares et autoimmunes, Centre Hospitalier de Mulhouse, Mulhouse, France
| | - Laurent Arnaud
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Ghada Alsaleh
- UMR 1109 Immuno-Rhumatologie Moléculaire, INSERM, Strasbourg, France
| | - François Lefebvre
- Department of Public Health, Hopitaux universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Vincent Gies
- UPR 3572 Immunopathology and Therapeutic Chemistry, CNRS, Strasbourg, France
| | - Thierry Martin
- UPR 3572 Immunopathology and Therapeutic Chemistry, CNRS, Strasbourg, France.,Department of Internal Medicine, Centre de références des maladies rares et auto immunes, Hopitaux universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Cédric Lukas
- Department of Rheumatology, Lapeyronie Hospital, Montpellier University, Montpellier, France
| | - Jean Durckel
- Department of Radiology, Hopitaux universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Marc Ardizzone
- Department of Rheumatology, Centre de compétence maladies rares et autoimmunes, Centre Hospitalier de Mulhouse, Mulhouse, France
| | - Rose-Marie Javier
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Jean-François Kleinmann
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Paul Moreau
- Department of Rheumatology, Hopital Louis Pasteur, Colmar, Alsace, France
| | - Gilles Blaison
- Department of Internal Medicine, Hopital Louis Pasteur, Colmar, Alsace, France
| | - Joelle Goetz
- Department of Immunology, Hopitaux universitaires de Strasbourg, Strasbourg, France
| | - Emmanuel Chatelus
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Jacques-Eric Gottenberg
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France.,UPR 3572 Immunopathology and Therapeutic Chemistry, CNRS, Strasbourg, France
| | - Jean Sibilia
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France.,UMR 1109 Immuno-Rhumatologie Moléculaire, INSERM, Strasbourg, France
| | - Christelle Sordet
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
13
|
Detrimental roles of TNF-alpha in the antiphospholipid syndrome and de novo synthesis of antiphospholipid antibodies induced by biopharmaceuticals against TNF-alpha. J Thromb Thrombolysis 2017; 44:565-570. [DOI: 10.1007/s11239-017-1571-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
14
|
Zhou J, Kawai T, Yu Q. Pathogenic role of endogenous TNF-α in the development of Sjögren's-like sialadenitis and secretory dysfunction in non-obese diabetic mice. J Transl Med 2017; 97:458-467. [PMID: 28067896 PMCID: PMC5376226 DOI: 10.1038/labinvest.2016.141] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/05/2016] [Accepted: 11/20/2016] [Indexed: 12/14/2022] Open
Abstract
Patients with Sjögren's syndrome (SS), an autoimmune disease primarily affecting exocrine glands, exhibit enhanced TNF-α expression in the saliva and salivary glands. However, the precise in vivo role of TNF-α during the initiation and development of SS is not clearly defined. The present study is undertaken to determine the function of endogenously produced TNF-α in the pathogenesis of SS in non-obese diabetic (NOD) mice, a model of this human disease. Administration of a neutralizing anti-TNF-α antibody to female NOD mice during the stage prior to disease onset significantly improved salivary secretion, indicating a remission of clinical symptoms of SS. TNF-α blockade also decreased the number of leukocyte foci and reduced the number of T cells and B cells in the submandibular glands (SMG). Moreover, TNF-α blockade reduced T-bet protein levels in the SMG, suggesting a decrease in T helper 1 and T cytotoxic 1 cells. These cellular changes induced by TNF-α neutralization were associated with a reduction in T- and B-cell chemoattractants CXCL9 and CXC13. In addition, TNF-α blockade markedly increased the expression level of tight junction protein claudin-1 and water channel protein aquaporin-5, two key factors required for normal salivary secretion, in the SMG. Collectively, these findings indicate that endogenous TNF-α has a pathogenic role in the development of SS in the NOD model of this disease.
Collapse
Affiliation(s)
| | | | - Qing Yu
- Address for correspondence and reprint requests: Corresponding Author: Qing Yu, M.D., Ph.D., Department of Immunology and Infectious Diseases, The Forsyth Institute, 245 First Street, Cambridge, MA 02142, Tel: 617-892-8310,
| |
Collapse
|
15
|
Parodis I, Ding H, Zickert A, Arnaud L, Larsson A, Svenungsson E, Mohan C, Gunnarsson I. Serum soluble tumour necrosis factor receptor-2 (sTNFR2) as a biomarker of kidney tissue damage and long-term renal outcome in lupus nephritis. Scand J Rheumatol 2016; 46:263-272. [PMID: 27973968 DOI: 10.1080/03009742.2016.1231339] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVES We investigated the performance of soluble tumour necrosis factor receptor-2 (sTNFR2) as a biomarker of renal activity, damage, treatment response, and long-term outcome in lupus nephritis (LN). METHOD Serum sTNFR2 levels were assessed in 64 LN patients (52 proliferative, 12 membranous) before and after induction treatment, and in 314 non-lupus controls. In LN patients, renal biopsies were performed at baseline and post-treatment. Patients with ≥ 50% reduced proteinuria, normal or improved estimated glomerular filtration rate (eGFR) by ≥ 25%, and inactive urinary sediment were considered clinical responders (CRs). Patients with ≥ 50% improved renal activity index were considered histopathological responders (HRs). Long-term renal outcome was determined using the chronic kidney disease (CKD) stage after a median follow-up of 11.3 years. RESULTS sTNFR2 levels were elevated in LN patients versus controls both at baseline (p < 0.001) and post-treatment (p < 0.001), and decreased following treatment (p < 0.001). Baseline sTNFR2 correlated with Chronicity Index scores in both baseline (r = 0.34, p = 0.006) and post-treatment (r = 0.43, p < 0.001) biopsies. In membranous LN, baseline sTNFR2 levels were higher in CRs (p = 0.048) and HRs (p = 0.03) than in non-responders, and decreased only in CRs (p = 0.03). Both baseline (p = 0.02) and post-treatment (p = 0.03) sTNFR2 levels were associated with decreasing eGFR throughout long-term follow-up, and post-treatment levels were higher in patients with long-term follow-up CKD stage ≥ 3 versus 1-2 (p = 0.008). CONCLUSIONS Our data suggest serum sTNFR2 as a marker of kidney tissue damage and a predictor of long-term prognosis in LN, and merit further evaluation of sTNFR2 as a predictor of clinical and histopathological treatment outcomes in membranous LN.
Collapse
Affiliation(s)
- I Parodis
- a Department of Medicine, Rheumatology Unit, Karolinska Institutet , Karolinska University Hospital , Stockholm , Sweden
| | - H Ding
- b Department of Biomedical Engineering , University of Houston , Houston , Texas , USA
| | - A Zickert
- a Department of Medicine, Rheumatology Unit, Karolinska Institutet , Karolinska University Hospital , Stockholm , Sweden
| | - L Arnaud
- a Department of Medicine, Rheumatology Unit, Karolinska Institutet , Karolinska University Hospital , Stockholm , Sweden
| | - A Larsson
- c Department of Medical Sciences/Clinical Chemistry , Uppsala University , Uppsala , Sweden
| | - E Svenungsson
- a Department of Medicine, Rheumatology Unit, Karolinska Institutet , Karolinska University Hospital , Stockholm , Sweden
| | - C Mohan
- b Department of Biomedical Engineering , University of Houston , Houston , Texas , USA
| | - I Gunnarsson
- a Department of Medicine, Rheumatology Unit, Karolinska Institutet , Karolinska University Hospital , Stockholm , Sweden
| |
Collapse
|
16
|
Birner P, Heider S, Petzelbauer P, Wolf P, Kornauth C, Kuroll M, Merkel O, Steiner G, Kishimoto T, Rose-John S, Soleiman A, Moriggl R, Kenner L. Interleukin-6 receptor alpha blockade improves skin lesions in a murine model of systemic lupus erythematosus. Exp Dermatol 2016; 25:305-10. [PMID: 26739431 PMCID: PMC4982049 DOI: 10.1111/exd.12934] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2015] [Indexed: 11/29/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease, characterized by antinuclear autoantibodies (ANA) and immunocomplexes, commonly affecting kidneys, skin, heart, lung or even the brain. We have shown that JunB(Δep) mice develop a SLE phenotype linked to increased epidermal Interleukin (IL)-6 secretion. Blocking of IL-6 receptor alpha (IL-6Rα) is considered as therapeutic strategy for the treatment of SLE. JunB(Δep) and wild-type mice were treated for short (5 weeks) or long term (21 weeks) with the IL-6Rα-blocking antibody MR16-1. Skin and kidney of mice were investigated by histology and immunofluorescence, and in addition, kidneys were analysed by electron microscopy. Furthermore, soluble IL-6R (sIL-6R), antihistone and antinucleosome antibodies levels were measured and associated with disease parameters. Treatment with MR16-1 resulted in significant improvement of SLE-like skin lesions in JunB(Δep) mice, compared to untreated mice. The sIL-6R amount upon long-term treatment with MR16-1 was significantly higher in JunB(Δep) versus untreated JunB(Δep) (P = 0.034) or wild-type mice (P = 0.034). MR16-1 treatment over these time spans did not significantly improve kidney pathology of immunoglobulin deposits causing impaired function. Significantly higher antihistone (P = 0.028) and antinucleosome antibody levels (P = 0.028) were measured in MR16-1-treated JunB(Δep) mice after treatment compared to levels before therapy. In conclusion, blockade of IL-6Rα improves skin lesions in a murine SLE model, but does not have a beneficial effect on autoimmune-mediated kidney pathology. Inhibition of IL-6R signalling might be helpful in lupus cases with predominant skin involvement, but combinatorial treatment might be required to restrain autoantibodies.
Collapse
Affiliation(s)
- Peter Birner
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Susanne Heider
- Department of Pathology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Peter Petzelbauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Peter Wolf
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
| | | | - Madeleine Kuroll
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Olaf Merkel
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Günter Steiner
- Division of Rheumatology, Department of Internal Medicine III, Osaka University, Suita City, Osaka, Japan
| | - Tadamitsu Kishimoto
- Laboratory of Immune Regulation, Graduate School of Frontier Biosciences, Osaka University, Suita City, Osaka, Japan
| | | | | | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria.,Medical University of Vienna, Vienna, Austria
| | - Lukas Kenner
- Department of Pathology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
17
|
Abstract
The bidirectional causality between kidney injury and inflammation remains an area of unexpected discoveries. The last decade unraveled the molecular mechanisms of sterile inflammation, which established danger signaling via pattern recognition receptors as a new concept of kidney injury-related inflammation. In contrast, renal cell necrosis remained considered a passive process executed either by the complement-related membrane attack complex, exotoxins, or cytotoxic T cells. Accumulating data now suggest that renal cell necrosis is a genetically determined and regulated process involving specific outside-in signaling pathways. These findings support a unifying theory in which kidney injury and inflammation are reciprocally enhanced in an autoamplification loop, referred to here as necroinflammation. This integrated concept is of potential clinical importance because it offers numerous innovative molecular targets for limiting kidney injury by blocking cell death, inflammation, or both. Here, the contribution of necroinflammation to AKI is discussed in thrombotic microangiopathies, necrotizing and crescentic GN, acute tubular necrosis, and infective pyelonephritis or sepsis. Potential new avenues are further discussed for abrogating necroinflammation-related kidney injury, and questions and strategies are listed for further exploration in this evolving field.
Collapse
Affiliation(s)
- Shrikant R Mulay
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany; and
| | - Andreas Linkermann
- Clinic for Nephrology and Hypertension, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Hans-Joachim Anders
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany; and
| |
Collapse
|
18
|
Pirowska MM, Goździalska A, Lipko-Godlewska S, Obtułowicz A, Sułowicz J, Podolec K, Wojas-Pelc A. Autoimmunogenicity during anti-TNF therapy in patients with psoriasis and psoriatic arthritis. Postepy Dermatol Alergol 2015; 32:250-4. [PMID: 26366147 PMCID: PMC4565839 DOI: 10.5114/pdia.2015.53320] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 04/28/2015] [Accepted: 04/30/2015] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION The tumor necrosis factor (TNF-α) was initially described as lymphotoxin or cachectin. The discovery of therapies blocking the action of TNF-α, in 1988, started a new era in the therapy. One of often reported adverse effects related to the use of TNF-α antagonists is induction of the formation of autologous antibodies and antibodies neutralizing anti-TNF drugs. The development of anti-TNF-induced lupus or classical drug-induced lupus is more rarely reported. AIM To evaluate the presence and the level of anti-nuclear antibodies in patients with psoriasis and psoriatic arthritis and the influence of anti-TNF therapy used on the concentration of antinuclear antibody (ANA). MATERIAL AND METHODS A total of 28 subjects were included in the study. 71.4% of subjects were diagnosed with psoriatic arthritis and 28.6% with plaque psoriasis. RESULTS Among the patients with plaque psoriasis, the antinuclear antibodies were found in 25% of subjects and in 80% of patients with psoriatic arthritis. After the treatment an increase in the titer or appearance of antibodies was found in 66.7% in the infliximab group, 18.2% in the etanercept group and 54.7% in the adalimumab group. No subjects developed symptoms of drug-induced systemic lupus. CONCLUSIONS Our findings have shown that all anti-TNF therapies induced ANA in psoriatic arthritis and psoriatic patients. Considering a mild course of lupus induced by anti-TNF treatment and, usually intrinsic, resolution of symptoms, the biological therapy still appears as a safe treatment for patients.
Collapse
Affiliation(s)
- Magdalena M. Pirowska
- Department of Dermatology, Jagiellonian University Medical College, Krakow, Poland. Head of the Department: Prof. Anna Wojas-Pelc MD, PhD
| | - Anna Goździalska
- Department of Health and Medical Science, Andrzej Frycz Modrzewski Krakow University, Krakow, Poland. Head of the Department: Prof. Jerzy Jaśkiewicz MD, PhD
| | - Sylwia Lipko-Godlewska
- Department of Dermatology, Jagiellonian University Medical College, Krakow, Poland. Head of the Department: Prof. Anna Wojas-Pelc MD, PhD
| | - Aleksander Obtułowicz
- Department of Dermatology, Jagiellonian University Medical College, Krakow, Poland. Head of the Department: Prof. Anna Wojas-Pelc MD, PhD
| | - Joanna Sułowicz
- Department of Dermatology, Jagiellonian University Medical College, Krakow, Poland. Head of the Department: Prof. Anna Wojas-Pelc MD, PhD
| | - Katarzyna Podolec
- Department of Dermatology, Jagiellonian University Medical College, Krakow, Poland. Head of the Department: Prof. Anna Wojas-Pelc MD, PhD
| | - Anna Wojas-Pelc
- Department of Dermatology, Jagiellonian University Medical College, Krakow, Poland. Head of the Department: Prof. Anna Wojas-Pelc MD, PhD
| |
Collapse
|
19
|
Cortés-Hernández J, Egri N, Vilardell-Tarrés M, Ordi-Ros J. Etanercept in refractory lupus arthritis: An observational study. Semin Arthritis Rheum 2015; 44:672-9. [PMID: 25712812 DOI: 10.1016/j.semarthrit.2015.01.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 12/22/2014] [Accepted: 01/16/2015] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To investigate the long-term safety and preliminary efficacy of etanercept in patients with refractory lupus arthritis. METHODS We evaluated 43 patients in this observational cohort study. All received etanercept (50mg/week) in addition to concomitant immunosuppressive agents. Patient and disease characteristics were collected. Incidence of adverse events and the effect on autoantibody levels were evaluated. Clinical efficacy was measured by the 28-joint count and the SLEDAI-2K scores. Remission of lupus arthritis was defined by a 28-joint score = 0. Clinically inactive systemic disease was defined by a SLEDAI-2K score <4. RESULTS The total follow-up time was 93 patient-years (median: 2.3 years per patient; range: 0.4-6.8 years). Most side effects were minor and related to local reactions. Only 2 significant adverse events occurred (8%), both were of infectious nature. The rate of autoantibody production was low (18%). A mild increase in titres of ANA (2), IgG anti-dsDNA (3) and IgM anticardiolipin (aCL) (2) antibodies was observed. All anti-dsDNA antibody increments were transient and coincided with systemic flares. No vascular events occurred. In general, disease activity declined during therapy. Most patients (83%) with lupus arthritis achieved clinical remission by week 12. All patients with simultaneous serositis experienced clinical and radiological resolution of this condition. Relapses were frequent (23%), mostly mild and related to etanercept reduction. A total of 24 patients discontinued treatment, 12 of them due to clinical remission. CONCLUSIONS Long-term therapy with etanercept was relatively safe and had remarkable long-term efficacy for refractory lupus arthritis. In view of these results, further controlled trials are warranted.
Collapse
Affiliation(s)
- Josefina Cortés-Hernández
- Systemic Autoimmune Diseases Unit, Internal Medicine Department, Universitari Vall d´Hebron Hospital, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Passeig Vall- d´Hebron 119-129, Barcelona 08035, Spain
| | - Natalia Egri
- Systemic Autoimmune Diseases Unit, Internal Medicine Department, Universitari Vall d´Hebron Hospital, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Passeig Vall- d´Hebron 119-129, Barcelona 08035, Spain
| | - Miquel Vilardell-Tarrés
- Systemic Autoimmune Diseases Unit, Internal Medicine Department, Universitari Vall d´Hebron Hospital, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Passeig Vall- d´Hebron 119-129, Barcelona 08035, Spain
| | - Josep Ordi-Ros
- Systemic Autoimmune Diseases Unit, Internal Medicine Department, Universitari Vall d´Hebron Hospital, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Passeig Vall- d´Hebron 119-129, Barcelona 08035, Spain.
| |
Collapse
|
20
|
Aringer M, Smolen JS. Safety of off-label biologicals in systemic lupus erythematosus. Expert Opin Drug Saf 2014; 14:243-51. [DOI: 10.1517/14740338.2015.986455] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
21
|
Karonitsch T, Aringer M. [Biologics in SLE]. Wien Med Wochenschr 2014; 165:40-5. [PMID: 25411010 DOI: 10.1007/s10354-014-0322-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 10/02/2014] [Indexed: 11/24/2022]
Abstract
Biologics have become indispensable in the last decade in the treatment of the more common rheumatic diseases. For treating systemic lupus erythematodes (SLE), B-cell depletion, albeit off-label, has been a well-accepted strategy in severe and refractory disease. Unexpectedly, however, the results of the first randomized controlled rituximab trials in SLE were negative. New trials with improved study protocols are ongoing, which should resolve this issue. In 2012, with the approval of belimumab, SLE finally entered the era of approved biological therapies. The anti-Blys/BAFF antibody belimumab showed prevention of SLE flares, glucocorticoid sparing, and significant improvement in the quality of life of SLE patients, in part by drastically reducing immune complex mediated fatigue. Positive reports on further targeting approaches give hope that additional biological agents will be available for SLE therapy soon.
Collapse
Affiliation(s)
- Thomas Karonitsch
- Klinische Abteilung für Rheumatologie, Universitätsklinik für Innere Medizin III, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090, Wien, Österreich,
| | | |
Collapse
|
22
|
Kolarz B, Majdan M, Darmochwal-Kolarz DA, Dryglewska M. Antiphospholipid antibodies during 6-month treatment with infliximab: a preliminary report. Med Sci Monit 2014; 20:1227-31. [PMID: 25027437 PMCID: PMC4109570 DOI: 10.12659/msm.890270] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The introduction of tumor necrosis factor (TNF) antagonists (adalimumab, infliximab, and etanercept) was a major advance and was highly important and beneficial in most rheumatoid arthritis (RA) patients. The adverse effects of this treatment are infrequent, but include opportunistic intracellular infection (especially the reactivation of latent Mycobacterium tuberculosis); exacerbation of demyelinating disorders; and the production of various types of antibodies such as antinuclear antibodies (ANA) or double-stranded DNA autoantibodies (dsDNA) and antiphospholipid antibodies (aPL) such as anti-cardiolipin antibodies (aCL) and anti-B2GP-I antibodies (B2GP-I). The aim of the study was to determine the prevalence of aCL and B2GP-I in IgM and IgG classes, using ELISA tests, during 6 months of follow-up in patients with refractory RA successfully treated with infliximab. MATERIAL/METHODS We determined the prevalence of aCL and B2GP-I in IgM and IgG classes, using ELISA tests, during 6 months of follow-up in patients with refractory RA successfully treated with infliximab. RESULTS We observed a statistically important increase only in the group of B2GP-I IgM (p<0.05). There are contradictory results concerning the ability of infliximab to induce aPL, but most authors confirm this phenomenon. CONCLUSIONS Further investigations are needed to determine if the new aPL appears in patients with β2-GPI gene polymorphisms such as leucine-to-valine substitution at position 247, which can lead to a conformational changes in β2-GPI protein, leading to aPL synthesis. The role of aPL in pathogenesis of APS is still unclear, but we should remember the immunogenic aspect of TNF antagonist treatment. Therefore, we recommend early detection of aPL and observation of the patient, paying special attention to signs and symptoms of thromboembolism.
Collapse
Affiliation(s)
- Bogdan Kolarz
- Department of Clinical Immunology, Medical University of Lublin, Lublin, Poland
| | - Maria Majdan
- Department of Rheumatology and Connective Tissue Diseases, Medical University of Lublin, Lublin, Poland
| | | | - Magdalena Dryglewska
- Department of Rheummatology and Connective Tissue Diseases, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
23
|
Inoue M, Shoda H, Seri Y, Kubo K, Kanda H, Fujio K, Yamamoto K. Three cases of lupus nephritis patients with serum interleukin-32γ detection. Lupus 2014; 23:1187-91. [PMID: 24879659 DOI: 10.1177/0961203314538108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE Interleukin-32 (IL-32) is an inflammatory cytokine that is associated with the pathogenesis of several connective tissue diseases. We measured serum IL-32γ concentrations of systemic lupus erythematosus (SLE) patients. METHODS Serum samples were obtained from SLE patients (n = 51), and healthy controls (n = 15). Serum IL-32 concentrations were measured using ELISA. Clinical information was obtained from medical records. RESULTS Serum IL-32γ was detectable in three cases of SLE patients, whereas it was not detected in any healthy controls. Case 1: a 44-year-old female with lupus nephritis (LN) (Class II) and antiphospholipid antibody syndrome. Serum IL-32γ was 5.1 pg/ml. Case 2: a 30-year-old female with a history of diffuse proliferative LN (Class IV G (A/C)) and pulmonary hemorrhage. Serum IL-32γ was 8.9 pg/ml. Case 3: a 45-year-old female with chronic LN. Serum IL-32γ was 9.1 pg/ml. All three cases of IL-32γ-detectable patients had histories of LN and one had an active disease. In the context of LN, serum IL-32γ was detectable in 18.8% (three of 16) of SLE patients with histories of LN. CONCLUSION We suppose that IL-32γ could contribute to the pathogenesis of renal diseases in some LN patients.
Collapse
Affiliation(s)
- M Inoue
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - H Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Y Seri
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - K Kubo
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - H Kanda
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - K Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - K Yamamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
24
|
Fanouriakis A, Boumpas DT, Bertsias GK. Balancing efficacy and toxicity of novel therapies in systemic lupus erythematosus. Expert Rev Clin Pharmacol 2014; 4:437-51. [DOI: 10.1586/ecp.11.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
25
|
Moura Filho JP, Peixoto RL, Martins LG, de Melo SD, de Carvalho LL, Pereira AKFDTC, Freire EAM. Lupus erythematosus: considerations about clinical, cutaneous and therapeutic aspects. An Bras Dermatol 2014; 89:118-25. [PMID: 24626656 PMCID: PMC3938362 DOI: 10.1590/abd1806-4841.20142146] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 01/18/2013] [Indexed: 12/31/2022] Open
Abstract
Systemic Lupus Erythematosus is a chronic inflammatory disease with multifactorial etiology. Although clinical manifestations are varied, the skin is an important target-organ, which contributes to the inclusion of skin lesions in 4 out of the 17 new criteria for the diagnosis of the disease, according to the Systemic Lupus International Collaborating Clinics. The cutaneous manifestations of lupus are pleomorphic. Depending on their clinical characteristics, they can be classified into Acute Cutaneous Lupus Erythematosus, Subacute Cutaneous Lupus Erythematosus, Chronic Cutaneous Lupus Erythematosus and Intermittent Cutaneous Lupus Erythematosus. Treatment is based on preventive measures, reversal of inflammation, prevention of damage to target organs and relief of adverse events due to pharmacological therapy. The most commonly used treatment options are topical, systemic and surgical treatment, as well as phototherapy. The correct handling of the cases depends on a careful evaluation of the morphology of the lesions and the patient's general status, always taking into consideration not only the benefits but also the side effects of each therapeutic proposal.
Collapse
Affiliation(s)
| | - Raiza Luna Peixoto
- Medical Students Paraiba Federal University (UFPB) - João Pessoa (PB),
Brazil
| | - Lívia Gomes Martins
- Medical Students Paraiba Federal University (UFPB) - João Pessoa (PB),
Brazil
| | | | | | | | | |
Collapse
|
26
|
Mediators of inflammation and their effect on resident renal cells: implications in lupus nephritis. Clin Dev Immunol 2013; 2013:317682. [PMID: 24171032 PMCID: PMC3793320 DOI: 10.1155/2013/317682] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 08/07/2013] [Indexed: 12/01/2022]
Abstract
Lupus nephritis affects up to 70% of patients with systemic lupus erythematosus and is a major cause of morbidity and mortality. It is characterized by a breakdown of immune tolerance, production of autoantibodies, and deposition of immune complexes within the kidney parenchyma, resulting in local inflammation and subsequent organ damage. To date, numerous mediators of inflammation have been implicated in the development and progression of lupus nephritis, and these include cytokines, chemokines, and glycosaminoglycans. Of these, type I interferons (IFNs) can increase both gene and protein expression of cytokines and chemokines associated with lupus susceptibility, and interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and hyaluronan have been shown to elicit both pro- and anti-inflammatory effects on infiltrating and resident renal cells depending on the status of their microenvironment. Expression of IL-6, TNF-α, type I IFNs, and hyaluronan are increased in the kidneys of patients and mice with active lupus nephritis and have been shown to contribute to disease pathogenesis. There is also evidence that despite clinical remission, ongoing inflammatory processes may occur within the glomerular and tubulointerstitial compartments of the kidney, which further promote kidney injury. In this review, we provide an overview of the synthesis and putative roles of IL-6, TNF-α, IFN-α, and hyaluronan in the pathogenesis of lupus nephritis focusing on their effects on human mesangial cells and proximal renal tubular epithelial cells.
Collapse
|
27
|
Takeuchi M. A systematic review of biologics for the treatment of noninfectious uveitis. Immunotherapy 2013; 5:91-102. [PMID: 23256801 DOI: 10.2217/imt.12.134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Noninfectious uveitis is a potentially sight-threatening ocular disorder and variable therapeutic strategies have been proposed. Biologic therapies were introduced as a new option for patients with uveitis refractory to the conventional therapy using corticosteroids and immunosuppressive agents, and 10 years have passed since the initiation. In this review, the author summarizes current articles on the assessment of therapeutic application of biologics for refractory uveitis including other autoimmune diseases. Although some results are based on investigation with insufficient clinical trials, especially in biologics, the majority of biologics indicate preferable outcomes on refractory uveitis, with remarkable promise to increase the possibility of long-term remissions.
Collapse
Affiliation(s)
- Masaru Takeuchi
- Department of Ophthalmology, National Defense Medical College, Saitama, Japan.
| |
Collapse
|
28
|
Aringer M, Günther C, Lee-Kirsch MA. Innate immune processes in lupus erythematosus. Clin Immunol 2013; 147:216-22. [DOI: 10.1016/j.clim.2012.11.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 11/23/2012] [Accepted: 11/24/2012] [Indexed: 11/30/2022]
|
29
|
Dang LJ, Lubel JS, Gunatheesan S, Hosking P, Su J. Drug-induced lupus and autoimmune hepatitis secondary to infliximab for psoriasis. Australas J Dermatol 2013; 55:75-9. [PMID: 23651182 DOI: 10.1111/ajd.12054] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2012] [Accepted: 02/18/2013] [Indexed: 12/16/2022]
Abstract
We describe a case of previously unreported autoimmune hepatitis and lupus-like syndrome induced by infliximab treatment for chronic plaque psoriasis. The condition resolved after withdrawal of infliximab, with the liver injury having been reversed and minimal periportal fibrosis. In a two-part discussion we review the current literature on the pharmacology of infliximab and provide recommendations for management of infliximab side effects.
Collapse
Affiliation(s)
- Lucy J Dang
- Dermatology Department, Monash University, Australia; Eastern Health Clinical School, Monash University, Australia
| | | | | | | | | |
Collapse
|
30
|
Postal M, Peliçari KO, Sinicato NA, Marini R, Costallat LTL, Appenzeller S. Th1/Th2 cytokine profile in childhood-onset systemic lupus erythematosus. Cytokine 2013; 61:785-91. [PMID: 23332615 DOI: 10.1016/j.cyto.2012.11.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 10/15/2012] [Accepted: 11/26/2012] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To determine the serum levels of Th1 (IL-12, IFN-γ,TNF-α) and Th2 (IL-5, IL-6 and IL-10) cytokines in childhood-onset SLE, first-degree relatives and healthy controls. To elucidate their association with disease activity, laboratory and treatment features. METHODS We included 60 consecutive childhood-onset SLE patients [median age 18 years (range 10-37)], 64 first-degree relatives [median 40 (range 28-52)] and 57 healthy [median age 19 years (range 6-30 years)] controls. Controls were age and sex-matched to SLE patients. SLE patients were assessed for clinical and laboratory SLE manifestations, disease activity (SLEDAI), damage (SDI) and current drug exposures. Mood and anxiety disorders were determined through Becks Depression (BDI) and Anxiety Inventory (BAI). Th1 (IL-12, IFN-γ,TNF-α) and Th2 (IL-5, IL-6 and IL-10) cytokines levels were measured by ELISA and compared by non-parametric tests. RESULTS Serum TNF-α (p=0.004), IL-6 (p=0.007) and IL-10 (p=0.03) levels were increased in childhood-onset SLE patients when compared to first-degree relatives and healthy controls. TNF-α levels were significantly increased in patients with active disease (p=0.014) and correlated directly with SLEDAI scores (r=0.39; p=0.002). IL-12 (p=0.042) and TNF-α (p=0.009) levels were significantly increased in patients with nephritis and TNF-α in patients with depression (p=0.001). No association between cytokine levels and SDI scores or medication was observed. CONCLUSION Th1 cytokines may play a role in the pathogenesis of neuropsychiatric and renal manifestations in childhood-onset SLE. The correlation with SLEDAI suggests that TNF-α may be a useful biomarker for disease activity in childhood-onset SLE, however longitudinal studies are necessary to determine if increase of this cytokine may predict flares in childhood-onset SLE.
Collapse
Affiliation(s)
- Mariana Postal
- Department of Medicine, Rheumatology Unit, Faculty of Medical Science, State University of Campinas, Brazil
| | | | | | | | | | | |
Collapse
|
31
|
Koenig KF, Groeschl I, Pesickova SS, Tesar V, Eisenberger U, Trendelenburg M. Serum cytokine profile in patients with active lupus nephritis. Cytokine 2012; 60:410-6. [PMID: 22846145 DOI: 10.1016/j.cyto.2012.07.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 06/14/2012] [Accepted: 07/02/2012] [Indexed: 11/26/2022]
Abstract
PURPOSE Determination of disease activity of lupus nephritis remains challenging. Since cytokines play a role as inflammatory mediators extending renal injury, measuring serum cytokine levels might help in the clinical assessment of patients with lupus nephritis. Therefore, the aim of this study was to determine the diagnostic value of a panel of serum cytokines in patients with active lupus nephritis. METHODS In this prospective controlled multicenter trial, sera of 12 patients with active lupus nephritis were collected in a clinical routine setting at the time of renal biopsy and 6 months afterwards. Fourteen patients with inactive systemic lupus erythematosus (SLE), and 14 healthy subjects were used as controls. Eleven cytokines (IL-4, IL-5, IL-6, IL-10, IL-12(p40), IL-12(p70), IL-18, TNF-α, TGF-β1, IFN-α2, IFN-γ) and two soluble receptors (IL-1ra and TNF-RII) were measured by cytokine multiplex assay. RESULTS In inactive SLE patients, serum levels of IL-10, IL-12(p40), IL-18 and TNF-RII were increased compared to healthy controls. Active lupus nephritis was found to be associated with further increase of these cytokine levels. Follow-up measurements in clinical remission of lupus nephritis showed downregulation of increased cytokines to levels found in inactive SLE. Most strikingly, TNF-RII serum level were elevated in all patients with active lupus nephritis (p<0.001) and declined after clinical remission (p<0.0005). CONCLUSION The cytokine multiplex assay used in our study allowed a fast and stable analysis of a panel of serum cytokines in a clinical routine setting. In addition, serum cytokines, especially TNF-RII, might be excellent markers of active lupus nephritis.
Collapse
|
32
|
Miyanishi M, Segawa K, Nagata S. Synergistic effect of Tim4 and MFG-E8 null mutations on the development of autoimmunity. Int Immunol 2012; 24:551-9. [PMID: 22723547 DOI: 10.1093/intimm/dxs064] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Phagocytes, including macrophages, recognize phosphatidylserine exposed on apoptotic cells as an "eat me" signal. Milk Fat Globule EGF Factor VIII (MFG-E8) is secreted by one subset of macrophages, whereas Tim4, a type I membrane protein, is expressed by another. These proteins bind tightly to phosphatidylserine on apoptotic cells and enhance their engulfment by macrophages. To study the contribution of these proteins to the engulfment of apoptotic cells, we established a mouse line that was deficient in the genes encoding MFG-E8 and Tim4. The null mutation of Tim4 impaired the ability of resident peritoneal macrophages, but not thioglycollate-elicited macrophages, to engulf apoptotic cells. Mice deficient in either MFG-E8 or Tim4 on the C57BL/6 background developed hardly any autoantibodies, but aged female mice deficient in both MFG-E8 and Tim4 developed autoantibodies in an age-dependent manner. Tumour necrosis factor (TNF) α is known to protect against systemic lupus erythematosus-type autoimmunity, whereas type I IFN accelerates the disease. Indeed, the administration of an anti-TNFα antibody or a reagent that stimulates the IFN-α production [2,6,10,14-tetramethylpentadecane (TMPD; also known as pristane)] enhanced the production of autoantibodies in the MFG-E8- and Tim4-double-deficient mice. These results suggest that the double deficiency of Tim4 and MFG-E8, phosphatidylserine-binding proteins, can trigger autoimmunity and that TNFα and type I IFN regulate reciprocally the development of autoimmune disease.
Collapse
Affiliation(s)
- Masanori Miyanishi
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Yoshida-Konoe, Sakyo, Japan
| | | | | |
Collapse
|
33
|
Abstract
Only after biological response modifiers have become available have we begun to understand some of the complex functions of TNF in the human immune system. TNF is clearly essential for fighting intracellular pathogens, but probably not essential for fighting tumors. TNF influence on the humoral immune response, in contrast, has been more complicated to decipher, since TNF blockade is associated with both autoantibody formation and (somewhat) reduced responses to vaccination. Novel data now show that TNF is good for the humoral immune response. Vaccinations still work, however, and should be strongly recommended.
Collapse
|
34
|
Aringer M, Smolen JS. Therapeutic blockade of TNF in patients with SLE—Promising or crazy? Autoimmun Rev 2012; 11:321-5. [PMID: 21619949 DOI: 10.1016/j.autrev.2011.05.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2011] [Indexed: 12/12/2022]
|
35
|
Kuhn A, Ruland V, Bonsmann G. Cutaneous lupus erythematosus: Update of therapeutic options. J Am Acad Dermatol 2011; 65:e195-213. [DOI: 10.1016/j.jaad.2010.06.017] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 05/28/2010] [Accepted: 06/04/2010] [Indexed: 12/23/2022]
|
36
|
Takakubo Y, Konttinen YT. Immune-regulatory mechanisms in systemic autoimmune and rheumatic diseases. Clin Dev Immunol 2011; 2012:941346. [PMID: 22110541 PMCID: PMC3207139 DOI: 10.1155/2012/941346] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 08/06/2011] [Accepted: 08/18/2011] [Indexed: 02/07/2023]
Abstract
Systemic autoimmune and rheumatic diseases (SAIRDs) are thought to develop due to the failure of autoimmune regulation and tolerance. Current therapies, such as biologics, have improved the clinical results of SAIRDs; however, they are not curative treatments. Recently, new discoveries have been made in immune tolerance and inflammation, such as tolerogenic dendritic cells, regulatory T and B cells, Th 17 cells, inflammatory and tolerogenic cytokines, and intracellular signaling pathways. They lay the foundation for the next generation of the therapies beyond the currently used biologic therapies. New drugs should target the core processes involved in disease mechanisms with the aim to attain complete cure combined with safety and low costs compared to the biologic agents. Re-establishment of autoimmune regulation and tolerance in SAIRDs by the end of the current decade should be the final and realistic target.
Collapse
Affiliation(s)
- Yuya Takakubo
- Department of Medicine, Biomedicum Helsinki, University of Helsinki, PO Box 700, Haartmaninkatu 8, 00029 HUS, Finland.
| | | |
Collapse
|
37
|
Pham T, Bachelez H, Berthelot JM, Blacher J, Bouhnik Y, Claudepierre P, Constantin A, Fautrel B, Gaudin P, Goëb V, Gossec L, Goupille P, Guillaume-Czitrom S, Hachulla E, Huet I, Jullien D, Launay O, Lemann M, Maillefert JF, Marolleau JP, Martinez V, Masson C, Morel J, Mouthon L, Pol S, Puéchal X, Richette P, Saraux A, Schaeverbeke T, Soubrier M, Sudre A, Tran TA, Viguier M, Vittecoq O, Wendling D, Mariette X, Sibilia J. TNF alpha antagonist therapy and safety monitoring. Joint Bone Spine 2011; 78 Suppl 1:15-185. [PMID: 21703545 DOI: 10.1016/s1297-319x(11)70001-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To develop and/or update fact sheets about TNFα antagonists treatments, in order to assist physicians in the management of patients with inflammatory joint disease. METHODS 1. selection by a committee of rheumatology experts of the main topics of interest for which fact sheets were desirable; 2. identification and review of publications relevant to each topic; 3. development and/or update of fact sheets based on three levels of evidence: evidence-based medicine, official recommendations, and expert opinion. The experts were rheumatologists and invited specialists in other fields, and they had extensive experience with the management of chronic inflammatory diseases, such as rheumatoid. They were members of the CRI (Club Rhumatismes et Inflammation), a section of the Société Francaise de Rhumatologie. Each fact sheet was revised by several experts and the overall process was coordinated by three experts. RESULTS Several topics of major interest were selected: contraindications of TNFα antagonists treatments, the management of adverse effects and concomitant diseases that may develop during these therapies, and the management of everyday situations such as pregnancy, surgery, and immunizations. After a review of the literature and discussions among experts, a consensus was developed about the content of the fact sheets presented here. These fact sheets focus on several points: 1. in RA and SpA, initiation and monitoring of TNFα antagonists treatments, management of patients with specific past histories, and specific clinical situations such as pregnancy; 2. diseases other than RA, such as juvenile idiopathic arthritis; 3. models of letters for informing the rheumatologist and general practitioner; 4. and patient information. CONCLUSION These TNFα antagonists treatments fact sheets built on evidence-based medicine and expert opinion will serve as a practical tool for assisting physicians who manage patients on these therapies. They will be available continuously at www.cri-net.com and updated at appropriate intervals.
Collapse
Affiliation(s)
- Thao Pham
- Rheumatology Department, CHU Sainte-Marguerite, Marseille, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
There are many new therapeutic directions for the disease systemic lupus erythematosus (SLE). Despite this, the US Food and Drug Administration (FDA) has approved only one biological agent and it involves B cells, now thought to play a significant role in the pathogenesis of SLE. The name of the drug is belimumab, which is an agent that removes the B-cell cytokine called B lymphocyte stimulation factor (BLyS). Rituximab did not achieve its primary endpoints, even though the consensus is that it may be effective in some forms of SLE including renal disease. The anticytokine therapies against interleukin (IL)-6, IL-10, IL-17 and tumor necrosis factor (TNF) are effective in their own ways and phase II and III trials are in progress. Of particular interest to immunologists are the anti-interferon alpha and gamma drugs, which show promise in the animal models. Modulation of costimulatory molecules; specifically, the anti CD40, CTLA-***Ig and ICOS/B7RP blockade agents offer possibilities for the future using new pathways heretofore limited to rheumatoid arthritis. Finally, the use of tyrosine kinase inhibitors is another direction that has been successful in the inhibition of SLE in the murine model; early trials in human SLE have begun.
Collapse
Affiliation(s)
- Wen Xiong
- Newark Beth Israel Medical Center — Rheumatology, Newark, New Jersey, USA
| | - Robert G. Lahita
- Newark Beth Israel Medical Center — Rheumatology, Newark, New Jersey, USA
| |
Collapse
|
39
|
Mikita N, Ikeda T, Ishiguro M, Furukawa F. Recent advances in cytokines in cutaneous and systemic lupus erythematosus. J Dermatol 2011; 38:839-49. [PMID: 21767292 DOI: 10.1111/j.1346-8138.2011.01237.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lupus erythematosus (LE) includes a broad spectrum of diseases from a cutaneous-limited type to a systemic type. Systemic lupus erythematosus (SLE) is a systemic autoimmune disease which affects multiple organs. Cutaneous lupus erythematosus (CLE) includes skin symptoms seen in SLE and cutaneous-limited LE. Although immune abnormalities, as well as heritable, hormonal and environmental factors, are involved in the pathology of LE, the actual pathogenesis is still unclear. Recently, the involvement of various cytokines has been shown in the pathogenesis of LE. Moreover, some trials with biological agents targeted specific cytokines are also ongoing for SLE. In this article, we review the contributions of major cytokines such as interferon, tumor necrosis factor-α and interleukin-18 to LE, especially SLE and CLE.
Collapse
Affiliation(s)
- Naoya Mikita
- Department of Dermatology, Wakayama Medical University, Wakayama Department of Dermatology, Hashimoto Municipal Hospital, Hashimoto, Japan.
| | | | | | | |
Collapse
|
40
|
Bethunaickan R, Berthier CC, Ramanujam M, Sahu R, Zhang W, Sun Y, Bottinger EP, Ivashkiv L, Kretzler M, Davidson A. A unique hybrid renal mononuclear phagocyte activation phenotype in murine systemic lupus erythematosus nephritis. THE JOURNAL OF IMMUNOLOGY 2011; 186:4994-5003. [PMID: 21411733 DOI: 10.4049/jimmunol.1003010] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Renal infiltration with mononuclear cells is associated with poor prognosis in systemic lupus erythematosus. A renal macrophage/dendritic cell signature is associated with the onset of nephritis in NZB/W mice, and immune-modulating therapies can reverse this signature and the associated renal damage despite ongoing immune complex deposition. In nephritic NZB/W mice, renal F4/80(hi)/CD11c(int) macrophages are located throughout the interstitium, whereas F4/80(lo)/CD11c(hi) dendritic cells accumulate in perivascular lymphoid aggregates. We show here that F4/80(hi)/CD11c(int) renal macrophages have a Gr1(lo)/Ly6C(lo)/VLA4(lo)/MHCII(hi)/CD43(lo)/CD62L(lo) phenotype different from that described for inflammatory macrophages. At nephritis onset, F4/80(hi)/CD11c(int) cells upregulate cell surface CD11b, acquire cathepsin and matrix metalloproteinase activity, and accumulate large numbers of autophagocytic vacuoles; these changes reverse after the induction of remission. Latex bead labeling of peripheral blood Gr1(lo) monocytes indicates that these are the source of F4/80(hi)/CD11c(int) macrophages. CD11c(hi)/MHCII(lo) dendritic cells are found in the kidneys only after proteinuria onset, turnover rapidly, and disappear rapidly after remission induction. Gene expression profiling of the F4/80(hi)/CD11c(int) population displays increased expression of proinflammatory, regulatory, and tissue repair/degradation-associated genes at nephritis onset that reverses with remission induction. Our findings suggest that mononuclear phagocytes with an aberrant activation profile contribute to tissue damage in lupus nephritis by mediating both local inflammation and excessive tissue remodeling.
Collapse
Affiliation(s)
- Ramalingam Bethunaickan
- Center for Autoimmunity and Musculoskeletal Diseases, Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Karampetsou MP, Liossis SNC, Sfikakis PP. TNF-α antagonists beyond approved indications: stories of success and prospects for the future. QJM 2010; 103:917-28. [PMID: 20802008 DOI: 10.1093/qjmed/hcq152] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Tumour necrosis factor alpha (TNF-α) is a key molecule of the inflammatory response and data derived from studies in experimental animal models and humans suggest that TNF-α may be implicated in the pathogenesis of various autoimmune and non-infectious inflammatory conditions. Over the past decade pharmaceutical agents directed against TNF-α (infliximab, adalimumab and etanercept) have been widely and successfully employed for the management of rheumatoid arthritis (RA), ankylosing spondylitis (AS), psoriasis, psoriatic arthritis, juvenile idiopathic arthritis and inflammatory bowel disease, whereas two novel anti-TNF-α agents, golimumab and certolimumab pegol, recently entered the market for the treatment of RA, AS, Crohn's disease and psoriasis. Encouraged by the positive results obtained from the use of TNF-α antagonists in terms of efficacy and safety and due to the increasingly accumulating evidence regarding the implication of TNF-α in the pathogenesis of numerous disorders, anti-TNF-α agents have been considered for the management of diseases other than the ones they were initially approved for. Although in the case of multiple sclerosis and chronic heart failure the outcome from the administration of TNF-α blockers had been less than favourable, in other cases of non-infectious inflammatory conditions the response to TNF-α inhibition had been fairly beneficial. More specifically, according to well-documented clinical trials, anti-TNF-α agents exhibited favourable results in Behçet's disease, non-infectious ocular inflammation, pyoderma gangrenosum and hidradenitis suppurativa. In this review we discuss the successful outcomes as well as the prospects for the future from the off-label use of TNF-α antagonists.
Collapse
Affiliation(s)
- M P Karampetsou
- Department of Medicine, University of Patras, 26504, Patras, Greece
| | | | | |
Collapse
|
42
|
Abstract
The dysfunctional immune response that characterizes systemic lupus erythematosus (SLE) associates with an unbalanced production of soluble mediators that are crucial in promoting and sustaining chronic inflammation. The successful use of biologics in several autoimmune diseases has led to studies in SLE aimed at contrasting the proinflammatory responses that contribute to tissue and organ damage in the disease. Several approaches have been developed and tested as potential therapeutic agents in SLE in preclinical studies and in clinical trials. This article provides an overview on antibody-based approaches in SLE that, although preliminary, have the potential to expand the current therapeutic possibilities in the disease.
Collapse
Affiliation(s)
- Antonio La Cava
- Division of Rheumatology, Department of Medicine, University of California Los Angeles, 1000 Veteran Avenue 32-59, Los Angeles, CA 90095-1670, USA.
| |
Collapse
|
43
|
Affiliation(s)
- L Arnaud
- Service de médecine interne-2, centre national de référence lupus systémique et syndrome des anticorps anti-phospholipides, hôpital Pitié-Salpêtrière, AP-HP, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | | |
Collapse
|
44
|
Abstract
Systemic lupus erythematosus is a prototype of heterogeneous autoimmune disease. There have been few newly approved therapeutic agents in lupus treatment for many reasons. Several animal studies and human data have shown that many potential cytokines are related to the pathogenesis and disease activity of systemic lupus erythematosus. Cytokines are produced by many immune cell types and have variable functions in the immune system. Following the success of biological agents in the treatment of inflammatory arthritis, inflammatory bowel disease, and psoriasis, biological targeting to specific cytokines or receptor molecules is now promising in the treatment of systemic lupus erythematosus. In addition to B-cell deleting modalities, clinical trials targeting potential cytokines associated with disease pathogenesis are underway at various clinical stages. Among potential cytokines, targeting agents against B-cell activating factor and interferon-alpha are in the most advanced stage, and belimumab (anti-B-cell activating factor antibody) could be the first biological agent approved in the treatment of systemic lupus erythematosus. Anti-tumor necrosis factor was tried with some success, but with a potential risk of infection in a small number of patients. In this review, we discuss the rationale for anticytokine therapies and review agents currently in clinical trials, and those that could be developed in the near future for systemic lupus erythematosus. We present the results mostly from published trials and data from http://clinicaltrials.gov/ct2/
Collapse
Affiliation(s)
- D-H. Yoo
- Division of Rheumatology, Hospital for Rheumatic Diseases, Institute of Rheumatology, Hanyang University College of Medicine, Seoul, Korea,
| |
Collapse
|
45
|
Accelerated atherosclerosis in systemic lupus erythematosus: role of proinflammatory cytokines and therapeutic approaches. J Biomed Biotechnol 2010; 2010. [PMID: 20936125 PMCID: PMC2948929 DOI: 10.1155/2010/607084] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 06/21/2010] [Accepted: 07/26/2010] [Indexed: 12/02/2022] Open
Abstract
Systemic lupus erythematosus (SLE), a chronic multisystem autoimmune disease with a broad range of clinical manifestations, is associated with accelerated atherosclerosis (AT) and increased risk of cardiovascular complications. Relevant factors directly influencing the development of AT comprise immune complex generation, complement activation, and changes in the production and activity of a complex network of cytokines, including type I and II interferons, B lymphocyte stimulator (BLyS), TNFα, IL-6, IL-17 and migration macrophage inhibitor (MIF). Autoantibodies, also responsible for cytokine expression and activation, play a supplementary key role in the development of AT. Genomic and proteomic studies have contributed to the discovery of genes and proteins involved in AT, including some that may be suitable to be used as biomarkers. All that data has allowed the development of new drugs, most of them evaluated in clinical trials: inhibitors of IFN and TNFα, B cell directed therapies, synthetic oligodeoxynucleotides, intravenous immunoglobulin, or statins. The focus of the present paper is to summarize recent evidence showing the role of cytokines in the development of AT in SLE and the rationale, and safety concerns, in the use of combined therapy to prevent AT and cardiovascular disease.
Collapse
|
46
|
Bazsó A, Sevcic K, Orbán I, Poór G, Balogh Z, Kiss E. Overlapping juvenile idiopathic arthritis and systemic lupus erythematosus: a case report. Rheumatol Int 2010; 31:695-8. [PMID: 20676642 DOI: 10.1007/s00296-010-1594-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2010] [Accepted: 07/14/2010] [Indexed: 11/26/2022]
Abstract
Hereby, we report the case of a 12-year-old girl developing oligoarthritis and progressing into a polyarticular form. Rheumatoid factor was positive, and juvenile idiopathic arthritis (JIA) was diagnosed. After a poor response to DMARDs, an anti-TNF agent (infliximab) was initiated, but to be discontinued due to an allergic reaction. The same complication was observed with the fully human derivative, adalimumab. At the age of 22, the patient presented septicemia with severe anemia and subsequent development of leukopenia, myocarditis with heart failure, and ANA, aSm, aSS-A, aCL positives, and nephrotic syndrome. These new clinical manifestations fulfilled the classification criteria for the diagnosis of systemic lupus erythematosus. Due to the poor therapeutic responses for both diseases, alternative medical options have to be considered, such as targeted therapy with anti-CD20 or interleukin-6 receptor antagonist monoclonal antibodies. This patient may also be a candidate for autologous hemopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Anna Bazsó
- National Institute of Rheumatology and Physiotherapy, Budapest, Hungary.
| | | | | | | | | | | |
Collapse
|
47
|
Greven D, Leng L, Bucala R. Autoimmune diseases: MIF as a therapeutic target. Expert Opin Ther Targets 2010; 14:253-64. [PMID: 20148714 DOI: 10.1517/14728220903551304] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IMPORTANCE OF THE FIELD Autoimmune inflammatory diseases occur commonly in developed countries. The treatment of these diseases is usually non-curative and is aimed at suppressing inflammatory end-organ damage. Macrophage migration inhibitory factor (MIF) is a multipotent cytokine that has been implicated in the pathogenesis of numerous autoimmune inflammatory disorders. The selective targeting of MIF with either anti-MIF antibody or specific MIF antagonists may offer new therapeutic avenues for these diseases. AREAS COVERED IN THIS REVIEW Our aim is to discuss MIF-directed therapies as a novel therapeutic approach. The review covers literature from the past 10 years. WHAT THE READER WILL GAIN MIF inhibition has been shown to be efficacious in many experimental and pre-clinical studies of autoimmune inflammatory diseases. The close regulatory relationship between MIF and glucocorticoids makes therapeutic antagonism of MIF a potential steroid-sparing therapy in patients with refractory autoimmune diseases. TAKE HOME MESSAGE We expect that MIF antagonism by either small-molecule- or antibody-based approaches will find wide application in the treatment of autoimmune inflammatory diseases. Such therapy also may be informed by the MIF genotype of affected patients.
Collapse
Affiliation(s)
- Dorothee Greven
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Meibergdreef 9, Amsterdam, Netherlands.
| | | | | |
Collapse
|
48
|
Cytokine networks in systemic lupus erythematosus. J Biomed Biotechnol 2010; 2010:676284. [PMID: 20414360 PMCID: PMC2857803 DOI: 10.1155/2010/676284] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 03/21/2010] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease more prominent in women and characterized by multiple organ damage. Imbalance in cytokine production and cytokine levels correlates with SLE progression, making the understanding of SLE cytokine networks very important for SLE treatment strategy and drug development. In this article, we review cytokine networks that may be involved in the pathogenesis of SLE by briefly describing abnormal cytokine production and serum cytokine levels in SLE patients. We also focus on the pathological roles of cytokines and their interactions in immunoregulatory networks and suggest how their disturbances may implicate in pathological conditions in SLE. Finally, we further discuss the influence of estrogen on these cytokine networks.
Collapse
|
49
|
Vereckei E, Kriván G, Réti M, Szodoray P, Poór G, Kiss E. Anti-TNF-alpha-induced anti-phospholipid syndrome manifested as necrotizing vasculitis. Scand J Rheumatol 2010; 39:175-7. [PMID: 20337548 DOI: 10.3109/03009740902832753] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
50
|
Abstract
The pharmacological management of systemic lupus erythematosus (SLE) is challenging owing to its unpredictable clinical course, the variable organ system involvement and the lack of clear understanding of disease pathogenesis. The widely used corticosteroids and immunosuppressive drugs, which can control disease activity, have serious, potentially fatal, side effects. In the last decade, a better understanding of lupus pathogenesis has led to the development of biological agents that are directed at biomarkers. However, these biologicals also exert side effects due to infections resulting from completely eliminating immune cells (e.g., B cells) or cytokine signals (e.g., interferon-alpha) or affecting molecular targets outside the immune system (CD40L on platelets). New biomarker-driven clinical trials are ongoing to evaluate the safety and efficacy of B-cell depletion, blocking of interferon signaling, inhibition of the mTOR pathway, and restoration of glutathione deficiency in lupus T cells.
Collapse
Affiliation(s)
- Lisa Francis
- Division of Rheumatology, Department of Medicine, SUNY, 750 East Adams Street, Syracuse, NY 13210, USA
| | | |
Collapse
|