51
|
Mansilla MJ, Navarro-Barriuso J, Presas-Rodríguez S, Teniente-Serra A, Quirant-Sánchez B, Ramo-Tello C, Martínez-Cáceres EM. Optimal response to dimethyl fumarate is mediated by a reduction of Th1-like Th17 cells after 3 months of treatment. CNS Neurosci Ther 2019; 25:995-1005. [PMID: 31066225 PMCID: PMC6698982 DOI: 10.1111/cns.13142] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 04/01/2019] [Accepted: 04/07/2019] [Indexed: 01/08/2023] Open
Abstract
Aim Dimethyl fumarate (DMF) is one of the most promising therapies for relapsing‐remitting multiple sclerosis (RRMS) patients since it has shown immunomodulatory and neuroprotective effects. However, a percentage of RRMS patients do not exhibit an optimal response to DMF. The objective of this study was to identify early biomarkers of treatment response by analyzing changes in peripheral leukocyte subpopulations directly in whole blood samples. Methods A longitudinal and prospective study analyzing peripheral blood leukocyte subpopulations in 22 RRMS patients before initiating DMF treatment (baseline) and at 1, 3, 6, and 12 months of follow‐up was performed. Differences between no evidence of disease activity (NEDA) and ongoing disease activity (ODA) patients were analyzed. Results The beneficial effect of DMF was associated with a specific depletion of memory CD4+ and CD8+ T lymphocytes and B cells. Importantly, only NEDA patients showed (a) a shift from a pro‐ to an antiinflammatory profile, with an increase of Th2 cells and a decrease of Th1‐like Th17 lymphocytes; and (b) an increase of regulatory CD56bright NK cells. Conclusion The optimal response to DMF is mediated by a shift to antiinflammatory and immunoregulatory profile, which puts forward Th1‐like Th17 lymphocytes as a potential early biomarker of treatment response.
Collapse
Affiliation(s)
- María José Mansilla
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Juan Navarro-Barriuso
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Silvia Presas-Rodríguez
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Badalona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Aina Teniente-Serra
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Bibiana Quirant-Sánchez
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Cristina Ramo-Tello
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Badalona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Eva María Martínez-Cáceres
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
52
|
Dickel H, Bruckner T, Höxtermann S, Dickel B, Trinder E, Altmeyer P. Fumaric acid ester-induced T-cell lymphopenia in the real-life treatment of psoriasis. J Eur Acad Dermatol Venereol 2019; 33:893-905. [PMID: 30680823 PMCID: PMC6593701 DOI: 10.1111/jdv.15448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 12/19/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Fumaric acid esters (FAEs) are used to treat psoriasis and are known to cause lymphopenia in roughly 60% of the patients. Much remains to be elucidated about the biological effects of FAEs on lymphocytes. OBJECTIVE To evaluate the influence of long-term FAE (Fumaderm® ) treatment on peripheral blood CD4+ and CD8+ T cells, CD19+ B cells and CD56+ natural killer (NK) cells in psoriasis. METHODS In this single-centre retrospective observational subcohort study, we obtained leucocyte and lymphocyte subset counts before initiating FAE therapy in 371 psoriasis patients (mean age, 47.8 years; 63.3% males) and monitored them during treatment (mean treatment duration, 2.9 years). Multiparametric flow cytometry was used for immunophenotyping. RESULTS FAEs significantly reduced the numbers of CD4+ T, CD8+ T, CD19+ B and CD56+ NK cells. Among lymphocyte subsets, the mean percentage reduction from baseline was always highest for CD8+ T cells, with a peak of 55.7% after 2 years of therapy. The risk of T-cell lymphopenia increased significantly with the age of the psoriasis patients at the time that FAE therapy was initiated. It was significantly decreased for the combination therapy with methotrexate and folic acid (vitamin B9) supplementation. Supporting evidence was found suggesting that T-cell lymphopenia enhances the effectiveness of FAE therapy. CONCLUSIONS Monitoring distinct T-cell subsets rather than just absolute lymphocyte counts may provide more meaningful insights into both the FAE treatment safety and efficacy. We therefore suggest optimizing pharmacovigilance by additionally monitoring CD4+ and CD8+ T-cell counts at regular intervals, especially in patients of middle to older age. Thus, further prospective studies are needed to establish evidence-based recommendations to guide dermatologists in the management of psoriasis patients who are taking FAEs and who develop low absolute T-cell counts.
Collapse
Affiliation(s)
- H. Dickel
- Department of Dermatology, Venereology and AllergologyRuhr University BochumBochumGermany
| | - T. Bruckner
- Institute of Medical Biometry and Informatics (IMBI)University Hospital HeidelbergHeidelbergGermany
| | - S. Höxtermann
- Department of Dermatology, Venereology and AllergologyRuhr University BochumBochumGermany
| | - B. Dickel
- Dermatology Practice Peter WenzelMDHattingenGermany
| | - E. Trinder
- Department of Dermatology, Venereology and AllergologyRuhr University BochumBochumGermany
| | - P. Altmeyer
- Department of Dermatology, Venereology and AllergologyRuhr University BochumBochumGermany
- Dermatology Practice at City ParkBochumGermany
| |
Collapse
|
53
|
Klotz L, Havla J, Schwab N, Hohlfeld R, Barnett M, Reddel S, Wiendl H. Risks and risk management in modern multiple sclerosis immunotherapeutic treatment. Ther Adv Neurol Disord 2019; 12:1756286419836571. [PMID: 30967901 PMCID: PMC6444778 DOI: 10.1177/1756286419836571] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 02/14/2019] [Indexed: 12/13/2022] Open
Abstract
In recent years, there has been a paradigm shift in the treatment of multiple
sclerosis (MS) owing to the approval of a number of new drugs with very distinct
mechanisms of action. All approved disease-modifying drugs primarily work
directly on the immune system. However, the identification of an ‘optimal
choice’ for individual patients with regard to treatment efficacy, treatment
adherence and side-effect profile has become increasingly complex including
conceptual as well as practical considerations. Similarly, there are
peculiarities and specific requirements with regard to treatment monitoring,
especially in relation to immunosuppression, the development of secondary
immune-related complications, as well as the existence of drug-specific on- and
off-target effects. Both classical immunosuppression and selective immune
interventions generate a spectrum of potential therapy-related complications.
This article provides a comprehensive overview of available immunotherapeutics
for MS and their risks, detailing individual mechanisms of action and
side-effect profiles. Furthermore, practical recommendations for patients
treated with modern MS immunotherapeutics are provided.
Collapse
Affiliation(s)
- Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University of Münster, Building A1, Albert Schweitzer Campus 1, 48149 Münster, Germany
| | - Joachim Havla
- Institute of Clinical Neuroimmunology, University Hospital; Data Integration for Future Medicine consortium (DIFUTURE), Ludwig-Maximilians University, Munich, Germany
| | - Nicholas Schwab
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University, Munich, Germany Munich Cluster for Systems Neurology, Ludwig-Maximilians University, Munich, Germany
| | | | - Stephen Reddel
- Brain and Mind Centre, University of Sydney, NSW, Australia
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University of Münster, Building A1, Albert Schweitzer Campus 1, 48149 Münster, Germany
| |
Collapse
|
54
|
Traub J, Traffehn S, Ochs J, Häusser-Kinzel S, Stephan S, Scannevin R, Brück W, Metz I, Weber MS. Dimethyl fumarate impairs differentiated B cells and fosters central nervous system integrity in treatment of multiple sclerosis. Brain Pathol 2019; 29:640-657. [PMID: 30706542 PMCID: PMC6849574 DOI: 10.1111/bpa.12711] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 01/28/2019] [Indexed: 12/13/2022] Open
Abstract
In multiple sclerosis (MS), the effect of dimethyl fumarate (DMF) treatment is primarily attributed to its capacity to dampen pathogenic T cells. Here, we tested whether DMF also modulates B cells, which are newly recognized key players in MS, and to which extent DMF restricts ongoing loss of oligodendrocytes and axons in the central nervous system (CNS). Therefore, blood samples and brain tissue from DMF-treated MS patients were analyzed by flow cytometry or histopathological examination, respectively. Complementary mechanistic studies were conducted in inflammatory as well as non-inflammatory CNS demyelinating mouse models. In this study, DMF reduced the frequency of antigen-experienced and memory B cells and rendered remaining B cells less prone to activation and production of pro-inflammatory cytokines. Dissecting the functional consequences of these alterations, we found that DMF ameliorated a B cell-accentuated experimental autoimmune encephalomyelitis model by diminishing the capacity of B cells to act as antigen-presenting cells for T cells. In a non-inflammatory model of toxic demyelination, DMF limited oligodendrocyte apoptosis, promoted maturation of oligodendrocyte precursors and reduced axonal damage. In a CNS biopsy of a DMF-treated MS patient, we equivalently observed higher numbers of mature oligodendrocytes as well as a reduced extent of axonal damage when compared to a cohort of treatment-naïve patients. In conclusion, we showed that besides suppressing T cells, DMF dampens pathogenic B cell functions, which probably contributes to its clinical effectiveness in relapsing MS. DMF treatment may furthermore limit chronically ongoing CNS tissue damage, which may reduce long-term disability in MS apart from its relapse-reducing capacity.
Collapse
Affiliation(s)
- Jan Traub
- Institute of Neuropathology, University Medical Center, Göttingen, Germany.,Department of Neurology, University Medical Center, Göttingen, Germany
| | - Sarah Traffehn
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Jasmin Ochs
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | | | - Schirin Stephan
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | | | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Imke Metz
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Martin S Weber
- Institute of Neuropathology, University Medical Center, Göttingen, Germany.,Department of Neurology, University Medical Center, Göttingen, Germany
| |
Collapse
|
55
|
Ntranos A, Ntranos V, Bonnefil V, Liu J, Kim-Schulze S, He Y, Zhu Y, Brandstadter R, Watson CT, Sharp AJ, Katz Sand I, Casaccia P. Fumarates target the metabolic-epigenetic interplay of brain-homing T cells in multiple sclerosis. Brain 2019; 142:647-661. [PMID: 30698680 PMCID: PMC6821213 DOI: 10.1093/brain/awy344] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/08/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023] Open
Abstract
Cell-permeable formulations of metabolites, such as fumaric acid esters, have been used as highly effective immunomodulators in patients with multiple sclerosis and yet their mechanism of action remains elusive. Since fumaric acid esters are metabolites, and cell metabolism is highly intertwined with the epigenetic regulation of gene expression, we investigated whether this metabolic-epigenetic interplay could be leveraged for therapeutic purposes. To this end we recruited 47 treatment-naïve and 35 fumaric acid ester-treated patients with multiple sclerosis, as well as 16 glatiramer acetate-treated patients as a non-metabolite treatment control. Here we identify a significant immunomodulatory effect of fumaric acid esters on the expression of the brain-homing chemokine receptor CCR6 in CD4 and CD8 T cells of patients with multiple sclerosis, which include T helper-17 and T cytotoxic-17 cells. We report differences in DNA methylation of CD4 T cells isolated from untreated and treated patients with multiple sclerosis, using the Illumina EPIC 850K BeadChip. We first demonstrate that Krebs cycle intermediates, such as fumaric acid esters, have a significantly higher impact on epigenome-wide DNA methylation changes in CD4 T cells compared to amino-acid polymers such as glatiramer acetate. We then define a fumaric acid ester treatment-specific hypermethylation effect on microRNA MIR-21, which is critical for the differentiation of T helper-17 cells. This hypermethylation effect was attributed to the subpopulation of T helper-17 cells using a decomposition analysis and was further validated in an independent prospective cohort of seven patients before and after treatment with fumaric acid esters. In vitro treatment of CD4 and CD8 T cells with fumaric acid esters supported a direct and dose-dependent effect on DNA methylation at the MIR-21 promoter. Finally, the upregulation of miR-21 transcripts and CCR6 expression was inhibited if CD4 or CD8 T cells stimulated under T helper-17 or T cytotoxic-17 polarizing conditions were treated with fumaric acid esters in vitro. These data collectively define a direct link between fumaric acid ester treatment and hypermethylation of the MIR-21 locus in both CD4 and CD8 T cells and suggest that the immunomodulatory effect of fumaric acid esters in multiple sclerosis is at least in part due to the epigenetic regulation of the brain-homing CCR6+ CD4 and CD8 T cells.
Collapse
Affiliation(s)
- Achilles Ntranos
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Valentina Bonnefil
- Neuroscience, Advanced Science Research Center at The Graduate Center of the City University of New York, New York, NY, USA
| | - Jia Liu
- Neuroscience, Advanced Science Research Center at The Graduate Center of the City University of New York, New York, NY, USA
| | - Seunghee Kim-Schulze
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ye He
- Neuroscience, Advanced Science Research Center at The Graduate Center of the City University of New York, New York, NY, USA
| | - Yunjiao Zhu
- Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Brandstadter
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Corey T Watson
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Andrew J Sharp
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ilana Katz Sand
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patrizia Casaccia
- Neuroscience, Advanced Science Research Center at The Graduate Center of the City University of New York, New York, NY, USA
- Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
56
|
Yadav SK, Soin D, Ito K, Dhib-Jalbut S. Insight into the mechanism of action of dimethyl fumarate in multiple sclerosis. J Mol Med (Berl) 2019; 97:463-472. [PMID: 30820593 DOI: 10.1007/s00109-019-01761-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 12/26/2022]
Abstract
Dimethyl fumarate (DMF) is an oral, disease-modifying agent for the treatment of relapsing-remitting multiple sclerosis (RRMS). However, details regarding its mode of action are still emerging. It is believed that the mode of action of DMF involves both nuclear factor erythroid-derived 2-related factor (Nrf2)-dependent and independent pathways, which lead to an anti-inflammatory immune response due to type II myeloid cell and Th2 cell differentiation and neuroprotection. In this review, we will focus on the molecular and signaling effects of DMF that lead to changes in peripheral immune cell composition and function, alteration in CNS cell-specific functions, and effect on the blood-brain barrier.
Collapse
Affiliation(s)
- Sudhir Kumar Yadav
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Devika Soin
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Kouichi Ito
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Suhayl Dhib-Jalbut
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
| |
Collapse
|
57
|
Scheu S, Ali S, Mann-Nüttel R, Richter L, Arolt V, Dannlowski U, Kuhlmann T, Klotz L, Alferink J. Interferon β-Mediated Protective Functions of Microglia in Central Nervous System Autoimmunity. Int J Mol Sci 2019; 20:E190. [PMID: 30621022 PMCID: PMC6337097 DOI: 10.3390/ijms20010190] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/23/2018] [Accepted: 12/28/2018] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) leading to demyelination and axonal damage. It often affects young adults and can lead to neurological disability. Interferon β (IFNβ) preparations represent widely used treatment regimens for patients with relapsing-remitting MS (RRMS) with therapeutic efficacy in reducing disease progression and frequency of acute exacerbations. In mice, IFNβ therapy has been shown to ameliorate experimental autoimmune encephalomyelitis (EAE), an animal model of MS while genetic deletion of IFNβ or its receptor augments clinical severity of disease. However, the complex mechanism of action of IFNβ in CNS autoimmunity has not been fully elucidated. Here, we review our current understanding of the origin, phenotype, and function of microglia and CNS immigrating macrophages in the pathogenesis of MS and EAE. In addition, we highlight the emerging roles of microglia as IFNβ-producing cells and vice versa the impact of IFNβ on microglia in CNS autoimmunity. We finally discuss recent progress in unraveling the underlying molecular mechanisms of IFNβ-mediated effects in EAE.
Collapse
Affiliation(s)
- Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany.
| | - Shafaqat Ali
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany.
- Department of Psychiatry and Psychotherapy, University of Münster, 48149 Münster, Germany.
- Cells in Motion, Cluster of Excellence, University of Münster, 48149 Münster, Germany.
| | - Ritu Mann-Nüttel
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany.
| | - Lisa Richter
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany.
| | - Volker Arolt
- Department of Psychiatry and Psychotherapy, University of Münster, 48149 Münster, Germany.
| | - Udo Dannlowski
- Department of Psychiatry and Psychotherapy, University of Münster, 48149 Münster, Germany.
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, 48149, Münster, Germany.
| | - Luisa Klotz
- Department of Neurology, University of Münster, 48149 Münster, Germany.
| | - Judith Alferink
- Department of Psychiatry and Psychotherapy, University of Münster, 48149 Münster, Germany.
- Cells in Motion, Cluster of Excellence, University of Münster, 48149 Münster, Germany.
| |
Collapse
|
58
|
Abstract
Multiple sclerosis treatment faces tremendous changes owing to the approval of new medications, some of which are available as oral formulations. Until now, the four orally available medications, fingolimod, dimethylfumarate (BG-12), teriflunomide, and cladribine have received market authorization, whereas laquinimod is still under development. Fingolimod is a sphingosine-1-phosphate inhibitor, which is typically used as escalation therapy and leads to up to 60% reduction of the annualized relapse rate, but might also have neuroprotective properties. In addition, there are three more specific S1P agonists in late stages of development: siponimod, ponesimod, and ozanimod. Dimethylfumarate has immunomodulatory and cytoprotective functions and is used as baseline therapy. Teriflunomide, the active metabolite of the rheumatoid arthritis medication leflunomide, targets the dihydroorotate dehydrogenase, thus inhibiting the proliferation of lymphocytes by depletion of pyrimidines. Here we will review the mechanisms of action, clinical trial data, as well as data about safety and tolerability of the compounds.
Collapse
Affiliation(s)
- Simon Faissner
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| |
Collapse
|
59
|
Narapureddy B, Dubey D. Clinical evaluation of dimethyl fumarate for the treatment of relapsing-remitting multiple sclerosis: efficacy, safety, patient experience and adherence. Patient Prefer Adherence 2019; 13:1655-1666. [PMID: 31631980 PMCID: PMC6778444 DOI: 10.2147/ppa.s187529] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/02/2019] [Indexed: 12/03/2022] Open
Abstract
Dimethyl fumarate (DMF) is an oral disease-modifying therapy approved for management of relapsing-remitting multiple sclerosis patients. Results from phase 3 clinical trials (DEFINE, CONFIRM) and follow-up study (ENDORSE) have provided good evidence for its efficacy and safety profile. Patient-reported outcomes (PROs) assessment revealed stabilization or boost in health-related quality of life and work productivity of patients treated with DMF compared to placebo reflecting a higher patient satisfaction to therapy. Being an oral agent with relatively favorable risk versus benefit profile DMF is commonly prescribed first-line agent. However, literature suggests that intolerance to side effects, especially gastrointestinal adverse effects and flushing is one of the major causes to compromised therapeutic compliance. An increase in the real-world incidence of progressive multifocal leukoencephalopathy and liver abnormality cases is also concerning. Several prevention and mitigation strategies like patient counseling, dose up-titration, pretreatment with aspirin, use of symptomatic therapy and frequent blood monitoring have demonstrated to be effective in tackling these adverse effects and promoting adherence to DMF. In this article, we review the efficacy, safety, PROs and patient adhere data, along with various measures to manage adverse events and promote compliance.
Collapse
Affiliation(s)
| | - Divyanshu Dubey
- Departments of Neurology Mayo Clinic, Rochester, MN, USA
- Laboratory Medicine and Pathology Mayo Clinic, Rochester, MN, USA
- Correspondence: Divyanshu DubeyDepartment of Laboratory Medicine & Pathology, and Neurology, 200 First Street S.W., Rochester, MN55905, USAEmail
| |
Collapse
|
60
|
Hosseini A, Masjedi A, Baradaran B, Hojjat‐Farsangi M, Ghalamfarsa G, Anvari E, Jadidi‐Niaragh F. Dimethyl fumarate: Regulatory effects on the immune system in the treatment of multiple sclerosis. J Cell Physiol 2018; 234:9943-9955. [DOI: 10.1002/jcp.27930] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 10/24/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Arezoo Hosseini
- Drug Applied Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Faculty of Medicine, Tabriz University of Medical Sciences Tabriz Iran
| | - Ali Masjedi
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Faculty of Medicine, Tabriz University of Medical Sciences Tabriz Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Faculty of Medicine, Tabriz University of Medical Sciences Tabriz Iran
| | - Mohammad Hojjat‐Farsangi
- Immune and Gene therapy Lab Department of Oncology‐Pathology Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute Stockholm Sweden
- Department of Immunology School of Medicine, Bushehr University of Medical Sciences Bushehr Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences Yasuj Iran
| | - Enayat Anvari
- Department of Physiology Faculty of Medicine, Ilam University of Medical Sciences Ilam Iran
| | - Farhad Jadidi‐Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Faculty of Medicine, Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
61
|
Montes Diaz G, Hupperts R, Fraussen J, Somers V. Dimethyl fumarate treatment in multiple sclerosis: Recent advances in clinical and immunological studies. Autoimmun Rev 2018; 17:1240-1250. [DOI: 10.1016/j.autrev.2018.07.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 07/11/2018] [Indexed: 12/30/2022]
|
62
|
Regulatory B and T lymphocytes in multiple sclerosis: friends or foes? AUTOIMMUNITY HIGHLIGHTS 2018; 9:9. [PMID: 30415321 PMCID: PMC6230324 DOI: 10.1007/s13317-018-0109-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
Abstract
Current clinical experience with immunomodulatory agents and monoclonal antibodies in principle has established the benefit of depleting lymphocytic populations in relapsing–remitting multiple sclerosis (RRMS). B and T cells may exert multiple pro-inflammatory actions, but also possess regulatory functions making their role in RRMS pathogenesis much more complex. There is no clear correlation of Tregs and Bregs with clinical features of the disease. Herein, we discuss the emerging data on regulatory T and B cell subset distributions in MS and their roles in the pathophysiology of MS and its murine model, experimental autoimmune encephalomyelitis (EAE). In addition, we summarize the immunomodulatory properties of certain MS therapeutic agents through their effect on such regulatory cell subsets and their relevance to clinical outcomes.
Collapse
|
63
|
Brück J, Glocova I, Geisel J, Kellerer C, Röcken M, Ghoreschi K. Dimethyl fumarate-induced IL-17low
IFN-γlow
IL-4+
Th cells protect mice from severe encephalomyelitis. Eur J Immunol 2018; 48:1588-1591. [DOI: 10.1002/eji.201747435] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 06/06/2018] [Accepted: 06/30/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Jürgen Brück
- Department of Dermatology; University Medical Center; Eberhard Karls University Tübingen; Tübingen Germany
| | - Ivana Glocova
- Department of Dermatology; University Medical Center; Eberhard Karls University Tübingen; Tübingen Germany
| | - Julia Geisel
- Department of Dermatology; University Medical Center; Eberhard Karls University Tübingen; Tübingen Germany
| | - Christina Kellerer
- Department of Dermatology; University Medical Center; Eberhard Karls University Tübingen; Tübingen Germany
| | - Martin Röcken
- Department of Dermatology; University Medical Center; Eberhard Karls University Tübingen; Tübingen Germany
| | - Kamran Ghoreschi
- Department of Dermatology; University Medical Center; Eberhard Karls University Tübingen; Tübingen Germany
| |
Collapse
|
64
|
Li R, Patterson KR, Bar-Or A. Reassessing B cell contributions in multiple sclerosis. Nat Immunol 2018; 19:696-707. [PMID: 29925992 DOI: 10.1038/s41590-018-0135-x] [Citation(s) in RCA: 283] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023]
Abstract
There is growing recognition that B cell contributions to normal immune responses extend well beyond their potential to become antibody-producing cells, including roles at the innate-adaptive interface and their potential to modulate the responses of other immune cells such as T cells and myeloid cells. These B cell functions can have both pathogenic and protective effects in the context of central nervous system (CNS) inflammation. Here, we review recent advances in the field of multiple sclerosis (MS), which has traditionally been viewed as primarily a T cell-mediated disease, and we consider antibody-dependent and, particularly, emerging antibody-independent functions of B cells that may be relevant in both the peripheral and CNS disease compartments.
Collapse
Affiliation(s)
- Rui Li
- Center for Neuroinflammation and Experimental Therapeutics (CNET) and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristina R Patterson
- Center for Neuroinflammation and Experimental Therapeutics (CNET) and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics (CNET) and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
65
|
Melnik BC, John SM, Chen W, Plewig G. T helper 17 cell/regulatory T-cell imbalance in hidradenitis suppurativa/acne inversa: the link to hair follicle dissection, obesity, smoking and autoimmune comorbidities. Br J Dermatol 2018; 179:260-272. [PMID: 29573406 DOI: 10.1111/bjd.16561] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Disintegration of the infundibula of terminal hair follicles (HFs) in intertriginous skin areas exhibits the histological hallmark of hidradenitis suppurativa (HS)/acne inversa, featuring a dissecting terminal hair folliculitis. Elevated serum levels of interleukin (IL)-17 and local increase in the ratio of proinflammatory T helper (Th)17 cells and anti-inflammatory regulatory T cells (Tregs) have been reported. Perifollicular Tregs play a key role in HF stem cell homeostasis and infundibular integrity. OBJECTIVES In this review, we evaluate the Th17/Treg ratio in HS, its aggravating conditions and associated comorbidities. Furthermore, we intended to clarify whether drugs with reported beneficial effects in the treatment of HS readjust the deviated Th17/Treg axis. METHODS PubMed-listed, peer-reviewed original research articles characterizing Th17/Treg regulation in HS/acne inversa and associated comorbidities were selected for this review. RESULTS This review presents HS as a disease that exhibits an increased Th17/Treg ratio. Perifollicular deficiencies in Treg numbers or function may disturb HF stem cell homeostasis, initiating infundibular dissection of terminal HFs and perifollicular inflammation. The Th17/Treg imbalance is aggravated by obesity, smoking and decreased Notch signalling. In addition, HS-associated autoimmune diseases exhibit a disturbed Th17/Treg axis resulting in a Th17-dominant state. All drugs that have beneficial effects in the treatment of HS normalize the Th17/Treg ratio. CONCLUSIONS HS immunopathogenesis is closely related to deviations of the Th17/Treg balance, which may negatively affect Treg-controlled HF stem cell homeostasis and infundibular integrity. Pharmacological intervention should not only attenuate Th17/IL-17 signalling, but should also improve Treg function in order to stabilize HF stem cell homeostasis and infundibular integrity.
Collapse
Affiliation(s)
- B C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Osnabrück, Germany
| | - S M John
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Osnabrück, Germany
| | - W Chen
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany
| | - G Plewig
- Department of Dermatology and Allergy, Ludwig-Maximilian-University of Munich, Munich, Germany
| |
Collapse
|
66
|
Cunill V, Massot M, Clemente A, Calles C, Andreu V, Núñez V, López-Gómez A, Díaz RM, Jiménez MDLR, Pons J, Vives-Bauzà C, Ferrer JM. Relapsing-Remitting Multiple Sclerosis Is Characterized by a T Follicular Cell Pro-Inflammatory Shift, Reverted by Dimethyl Fumarate Treatment. Front Immunol 2018; 9:1097. [PMID: 29896193 PMCID: PMC5986897 DOI: 10.3389/fimmu.2018.01097] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/02/2018] [Indexed: 12/22/2022] Open
Abstract
Multiple sclerosis (MS) is considered a T cell-mediated autoimmune disease, although several evidences also demonstrate a B cell involvement in its etiology. Follicular T helper (Tfh) cells, a CXCR5-expressing CD4+ T cell subpopulation, are essential in the regulation of B cell differentiation and maintenance of humoral immunity. Alterations in circulating (c)Tfh distribution and/or function have been associated with autoimmune diseases including MS. Dimethyl fumarate (DMF) is a recently approved first-line treatment for relapsing-remitting MS (RRMS) patients whose mechanism of action is not completely understood. The aim of our study was to compare cTfh subpopulations between RRMS patients and healthy subjects and evaluate the impact of DMF treatment on these subpopulations, relating them to changes in B cells and humoral response. We analyzed, by flow cytometry, the distribution of cTfh1 (CXCR3+CCR6-), cTfh2 (CXCR3-CCR6-), cTfh17 (CXCR3-CCR6+), and the recently described cTfh17.1 (CXCR3+CCR6+) subpopulations of CD4+ Tfh (CD45RA-CXCR5+) cells in a cohort of 29 untreated RRMS compared to healthy subjects. CD4+ non-follicular T helper (Th) cells (CD45RA-CXCR5-) were also studied. We also evaluated the effect of DMF treatment on these subpopulations after 6 and 12 months treatment. Untreated RRMS patients presented higher percentages of cTfh17.1 cells and lower percentages of cTfh2 cells consistent with a pro-inflammatory bias compared to healthy subjects. DMF treatment induced a progressive increase in cTfh2 cells, accompanied by a decrease in cTfh1 and the pathogenic cTfh17.1 cells. A similar decrease of non-follicular Th1 and Th17.1 cells in addition to an increase in the anti-inflammatory Th2 subpopulation were also detected upon DMF treatment, accompanied by an increase in naïve B cells and a decrease in switched memory B cells and serum levels of IgA, IgG2, and IgG3. Interestingly, this effect was not observed in three patients in whom DMF had to be discontinued due to an absence of clinical response. Our results demonstrate a possibly pathogenic cTfh pro-inflammatory profile in RRMS patients, defined by high cTfh17.1 and low cTfh2 subpopulations that is reverted by DMF treatment. Monitoring cTfh subsets during treatment may become a biological marker of DMF effectiveness.
Collapse
Affiliation(s)
- Vanesa Cunill
- Immunology Department, Hospital Universitari Son Espases, Palma, Spain.,Human Immunopathology Research Laboratory, Institut d'Investigació Sanitària de les Illes Balears (IdISBa), Palma, Spain
| | - Margarita Massot
- Neurology Department, Hospital Universitari Son Espases, Palma, Spain
| | - Antonio Clemente
- Human Immunopathology Research Laboratory, Institut d'Investigació Sanitària de les Illes Balears (IdISBa), Palma, Spain.,Clinical Trials and Methodology Support Platform, Institut d'Investigació Sanitària de les Illes Balears (IdISBa), Palma, Spain
| | - Carmen Calles
- Neurology Department, Hospital Universitari Son Espases, Palma, Spain
| | - Valero Andreu
- Human Immunopathology Research Laboratory, Institut d'Investigació Sanitària de les Illes Balears (IdISBa), Palma, Spain
| | - Vanessa Núñez
- Neurology Department, Hospital Universitari Son Espases, Palma, Spain
| | - Antonio López-Gómez
- Immunology Department, Hospital Universitari Son Espases, Palma, Spain.,Human Immunopathology Research Laboratory, Institut d'Investigació Sanitària de les Illes Balears (IdISBa), Palma, Spain
| | - Rosa María Díaz
- Neurology Department, Hospital Universitari Son Espases, Palma, Spain
| | - María de Los Reyes Jiménez
- Immunology Department, Hospital Universitari Son Espases, Palma, Spain.,Human Immunopathology Research Laboratory, Institut d'Investigació Sanitària de les Illes Balears (IdISBa), Palma, Spain
| | - Jaime Pons
- Immunology Department, Hospital Universitari Son Espases, Palma, Spain.,Human Immunopathology Research Laboratory, Institut d'Investigació Sanitària de les Illes Balears (IdISBa), Palma, Spain
| | - Cristòfol Vives-Bauzà
- Research Unit, Institut d'Investigació Sanitària de les Illes Balears and Hospital Universitari Son Espases, Palma, Spain
| | - Joana Maria Ferrer
- Immunology Department, Hospital Universitari Son Espases, Palma, Spain.,Human Immunopathology Research Laboratory, Institut d'Investigació Sanitària de les Illes Balears (IdISBa), Palma, Spain
| |
Collapse
|
67
|
Dimethyl fumarate induces a persistent change in the composition of the innate and adaptive immune system in multiple sclerosis patients. Sci Rep 2018; 8:8194. [PMID: 29844361 PMCID: PMC5974280 DOI: 10.1038/s41598-018-26519-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/14/2018] [Indexed: 11/22/2022] Open
Abstract
The effects of dimethyl fumarate (DMF) on the immune system in multiple sclerosis (MS) are not completely elucidated. In this study, an extensive immunophenotypic analysis of innate and adaptive immune cells of DMF-treated MS patients was performed. Peripheral blood immune cell phenotypes were determined using flow cytometry in a follow-up study of 12 MS patients before, after 3 and 12 months of DMF treatment and a cross-sectional study of 25 untreated and 64 DMF-treated MS patients. Direct effects of DMF on B cells were analyzed in vitro. After 12 months of DMF treatment, percentages of monocytes, natural killer cells, naive T and B cells and transitional B cells increased. Percentages of (effector) memory T cells, (non) class-switched memory B cells and double negative B cells decreased together with CD4+ T cells expressing interferon-γ (IFN-γ), granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin-17 (IL-17). DMF treatment was fully effective as of 6 months and directly induced apoptosis and decreased expression of costimulatory CD40, antigen presentation molecule MHCII and B cell activating factor receptor (BAFFR) on B cells. DMF induced a persistent change of the immune system of MS patients, directly induced apoptosis and reduced expression of functional markers on B cells.
Collapse
|
68
|
Dimethyl fumarate downregulates the immune response through the HCA 2/GPR109A pathway: Implications for the treatment of multiple sclerosis. Mult Scler Relat Disord 2018; 23:46-50. [PMID: 29763776 DOI: 10.1016/j.msard.2018.04.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/02/2018] [Accepted: 04/21/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND The mechanisms of action of dimethyl fumarate (DMF), and its metabolite, monomethyl fumarate (MMF), for the treatment of multiple sclerosis are not completely elucidated. OBJECTIVES To discuss the role of DMF/MMF-induced hydroxycarboxylic acid receptor 2 (HCA2/GPR109A) pathway activation in the immune response and treatment of MS. METHODS A narrative (traditional) review of the current literature. RESULTS Studies have shown that binding of DMF/MMF to HCA2 on dendritic cells inhibits the production of pro-inflammatory cytokines in vitro and in MS murine models. Evidence suggests that activation of HCA2 expressed in immune cells and gut epithelial cells by DMF/MMF, may induce anti-inflammatory responses in the intestinal mucosa. CONCLUSION Although the DMF/MMF mechanism of action remains unclear, evidence suggests that the activation of HCA2/GPR109A pathway downregulates the immune response and may activate anti-inflammatory response in the intestinal mucosa, possibly leading to reduction in CNS tissue damage in MS patients.
Collapse
|
69
|
Brück J, Dringen R, Amasuno A, Pau-Charles I, Ghoreschi K. A review of the mechanisms of action of dimethylfumarate in the treatment of psoriasis. Exp Dermatol 2018; 27:611-624. [DOI: 10.1111/exd.13548] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Jürgen Brück
- Department of Dermatology; University Medical Center; Eberhard Karls University; Tübingen Germany
| | - Ralf Dringen
- Faculty 2 (Biology/Chemistry); Center for Biomolecular Interactions Bremen; University of Bremen; Bremen Germany
- Center for Environmental Research and Sustainable Technology; University of Bremen; Bremen Germany
| | | | | | - Kamran Ghoreschi
- Department of Dermatology; University Medical Center; Eberhard Karls University; Tübingen Germany
| |
Collapse
|
70
|
Kornberg MD, Bhargava P, Kim PM, Putluri V, Snowman AM, Putluri N, Calabresi PA, Snyder SH. Dimethyl fumarate targets GAPDH and aerobic glycolysis to modulate immunity. Science 2018; 360:449-453. [PMID: 29599194 DOI: 10.1126/science.aan4665] [Citation(s) in RCA: 488] [Impact Index Per Article: 69.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 01/29/2018] [Accepted: 03/15/2018] [Indexed: 12/27/2022]
Abstract
Activated immune cells undergo a metabolic switch to aerobic glycolysis akin to the Warburg effect, thereby presenting a potential therapeutic target in autoimmune disease. Dimethyl fumarate (DMF), a derivative of the Krebs cycle intermediate fumarate, is an immunomodulatory drug used to treat multiple sclerosis and psoriasis. Although its therapeutic mechanism remains uncertain, DMF covalently modifies cysteine residues in a process termed succination. We found that DMF succinates and inactivates the catalytic cysteine of the glycolytic enzyme glyceraldehyde 3-phosphate dehydrogenase (GAPDH) in mice and humans, both in vitro and in vivo. It thereby down-regulates aerobic glycolysis in activated myeloid and lymphoid cells, which mediates its anti-inflammatory effects. Our results provide mechanistic insight into immune modulation by DMF and represent a proof of concept that aerobic glycolysis is a therapeutic target in autoimmunity.
Collapse
Affiliation(s)
- Michael D Kornberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Paul M Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Vasanta Putluri
- Advanced Technology Core, Baylor College of Medicine, Houston, TX 77030, USA
| | - Adele M Snowman
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nagireddy Putluri
- Advanced Technology Core, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Solomon H Snyder
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. .,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
71
|
Abstract
Multiple sclerosis treatment faces tremendous changes as a result of the approval of new medications. The new medications have differing safety considerations and risks after long-term treatment, which are important for treating physicians to optimize and individualize multiple sclerosis care. Since the approval of the first multiple sclerosis capsule, fingolimod, the armamentarium of multiple sclerosis therapy has grown with the orally available medications dimethyl fumarate and teriflunomide. Fingolimod is mainly associated with cardiac side effects, dimethyl fumarate with bowel symptoms. Several reports about progressive multifocal leukoencephalopathy as a result of dimethyl fumarate or fingolimod therapy raised the awareness of fatal opportunistic infections. Alemtuzumab, a CD52-depleting antibody, is highly effective in reducing relapses but leads to secondary immunity with mainly thyroid disorders in about 30% of patients. Development of secondary B-cell-mediated disease might also be a risk of this antibody. The follow-up drug of the B-cell-depleting antibody rituximab, ocrelizumab, is mainly associated with infusion-related reactions; long-term data are scarce. The medication daclizumab high yield process, acting via the activation of CD56bright natural killer cells, can induce the elevation of liver function enzymes, but also fulminant liver failure has been reported. Therefore, daclizumab has been retracted from the market. Long-term data on the purine nucleoside cladribine in MS therapy, recently authorized in the European Union, have been acquired during the long-term follow-up of the cladribine studies. The small molecule laquinimod is currently under development. We review data of clinical trials and their extensions regarding long-term efficacy and side effects, which might be associated with long-term treatment.
Collapse
Affiliation(s)
- Simon Faissner
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstr. 56, Bochum, 44791, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstr. 56, Bochum, 44791, Germany.
| |
Collapse
|
72
|
Mills EA, Mao-Draayer Y. Understanding Progressive Multifocal Leukoencephalopathy Risk in Multiple Sclerosis Patients Treated with Immunomodulatory Therapies: A Bird's Eye View. Front Immunol 2018; 9:138. [PMID: 29456537 PMCID: PMC5801425 DOI: 10.3389/fimmu.2018.00138] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/16/2018] [Indexed: 12/14/2022] Open
Abstract
The increased use of newer potent immunomodulatory therapies for multiple sclerosis (MS), including natalizumab, fingolimod, and dimethyl fumarate, has expanded the patient population at risk for developing progressive multifocal leukoencephalopathy (PML). These MS therapies shift the profile of lymphocytes within the central nervous system (CNS) leading to increased anti-inflammatory subsets and decreased immunosurveillance. Similar to MS, PML is a demyelinating disease of the CNS, but it is caused by the JC virus. The manifestation of PML requires the presence of an active, genetically rearranged form of the JC virus within CNS glial cells, coupled with the loss of appropriate JC virus-specific immune responses. The reliability of metrics used to predict risk for PML could be improved if all three components, i.e., viral genetic strain, localization, and host immune function, were taken into account. Advances in our understanding of the critical lymphocyte subpopulation changes induced by these MS therapies and ability to detect viral mutation and reactivation will facilitate efforts to develop these metrics.
Collapse
Affiliation(s)
- Elizabeth A Mills
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States.,Graduate Program in Immunology, Program in Biomedical Sciences, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
73
|
Smith MD, Calabresi PA, Bhargava P. Dimethyl fumarate treatment alters NK cell function in multiple sclerosis. Eur J Immunol 2018; 48:380-383. [PMID: 29108094 PMCID: PMC7577262 DOI: 10.1002/eji.201747277] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/10/2017] [Accepted: 11/02/2017] [Indexed: 01/08/2023]
Abstract
Dimethyl fumarate (DMF) treatment in multiple sclerosis (MS) increased the proportion of immunoregulatory CD56bright NK cells and this change was proportional to reductions in CD8+ memory T cells. DMF and monomethyl fumarate (MMF) treatment in vitro had directs effects on NK cells and promoted degranulation and cytotoxicity.
Collapse
Affiliation(s)
- Matthew D Smith
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins University, Baltimore, MD, USA
| | - Peter A Calabresi
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins University, Baltimore, MD, USA
| | - Pavan Bhargava
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
74
|
Male-specific IL-33 expression regulates sex-dimorphic EAE susceptibility. Proc Natl Acad Sci U S A 2018; 115:E1520-E1529. [PMID: 29378942 DOI: 10.1073/pnas.1710401115] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The cellular and molecular basis of sex-dimorphic autoimmune diseases, such as the CNS demyelinating disease multiple sclerosis (MS), remains unclear. Our studies in the SJL mouse model of MS, experimental autoimmune encephalomyelitis (EAE), reveal that sex-determined differences in Il33 expression by innate immune cells in response to myelin peptide immunization regulate EAE susceptibility. IL-33 is selectively induced in PLP139-151-immunized males and activates type 2 innate lymphoid cells (ILC2s), cells that promote and sustain a nonpathogenic Th2 myelin-specific response. Without this attenuating IL-33 response, females generate an encephalitogenic Th17-dominant response, which can be reversed by IL-33 treatment. Mast cells are one source of IL-33 and we provide evidence that testosterone directly induces Il33 gene expression and also exerts effects on the potential for Il33 gene expression during mast cell development. Thus, in contrast to their pathogenic role in allergy, we propose a sex-specific role for both mast cells and ILC2s as attenuators of the pathogenic Th response in CNS inflammatory disease.
Collapse
|
75
|
Mills EA, Ogrodnik MA, Plave A, Mao-Draayer Y. Emerging Understanding of the Mechanism of Action for Dimethyl Fumarate in the Treatment of Multiple Sclerosis. Front Neurol 2018; 9:5. [PMID: 29410647 PMCID: PMC5787128 DOI: 10.3389/fneur.2018.00005] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/03/2018] [Indexed: 01/22/2023] Open
Abstract
Dimethyl fumarate (DMF) is an effective treatment option for relapsing-remitting multiple sclerosis (MS), but its therapeutic mechanism of action has not been fully elucidated. A better understanding of its mechanism will allow for the development of assays to monitor its clinical efficacy and safety in patients, as well as guide the development of the next generation of therapies for MS. In order to build the foundation for determining its mechanism, we reviewed the manner in which DMF alters lymphocyte subsets in MS patients, its impact on clinical efficacy and safety, as well as its molecular effects in cellular and animal models. DMF decreases absolute lymphocyte counts, but does not affect all subsets uniformly. CD8+ T-cells are the most profoundly affected, but reduction also occurs in the CD4+ population, particularly within the pro-inflammatory T-helper Th1 and Th17 subsets, creating a bias toward more anti-inflammatory Th2 and regulatory subsets. Similarly, B-lymphocyte, myeloid, and natural killer populations are also shifted toward a more anti-inflammatory state. In vitro and animal models demonstrate a role for DMF within the central nervous system (CNS) in promoting neuronal survival in an Nrf2 pathway-dependent manner. However, the impact of DMF directly within the CNS of MS patients remains largely unknown.
Collapse
Affiliation(s)
- Elizabeth A Mills
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Magdalena A Ogrodnik
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrew Plave
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States.,Graduate Program in Immunology, Program in Biomedical Sciences, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
76
|
Mrowietz U, Morrison PJ, Suhrkamp I, Kumanova M, Clement B. The Pharmacokinetics of Fumaric Acid Esters Reveal Their In Vivo Effects. Trends Pharmacol Sci 2018; 39:1-12. [DOI: 10.1016/j.tips.2017.11.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/06/2017] [Accepted: 11/08/2017] [Indexed: 12/21/2022]
|
77
|
Nakhaei-Nejad M, Barilla D, Lee CH, Blevins G, Giuliani F. Characterization of lymphopenia in patients with MS treated with dimethyl fumarate and fingolimod. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2017; 5:e432. [PMID: 29296636 PMCID: PMC5746425 DOI: 10.1212/nxi.0000000000000432] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/03/2017] [Indexed: 01/08/2023]
Abstract
Objective: Lymphopenia is a common occurrence of disease-modifying therapies (DMTs) for relapsing-remitting MS (RRMS). The aim of this study was to dissect the prevalence of various lymphocyte subsets in patients with RRMS treated with 2 DMTs commonly associated with lymphopenia, dimethyl fumarate (DMF), and fingolimod (FTY). Methods: Multicolor flow cytometry and multiplex assays were used to identify up to 50 lymphocyte subpopulations and to examine the expression of multiple cytokines in selected patients. We compared patients untreated (NT) or treated with FTY or DMF who did (DMF-L) or did not (DMF-N) develop lymphopenia. Results: All FTY patients developed lymphopenia in both T-cell and B-cell compartments. CD41 T cells were more affected by this treatment than CD81 cells. In the B-cell compartment, the CD271IgD2 subpopulation was reduced. T cells but not B cells were significantly reduced in DMF-L. However, within the B cells, CD271 cells were significantly lower. Both CD41 and CD81 subpopulations were reduced in DMF-L. Within the remaining CD41 and CD81 compartments, there was an expansion of the naive subpopulation and a reduction of the effector memory subpopulation. Unactivated lymphocyte from DMF-L patients had significantly higher levels of interferon-γ, interleukin (IL)-12, IL-2, IL-4, IL-6, and IL-1β compared with DMF-N. In plasma, TNFβ was significantly higher in DMF-N and DMF-L compared with NT, whereas CCL17 was significantly higher in DMF-L compared with NT and DMF-N. Conclusions: This study shows that different treatments can target different lymphocyte compartments and suggests that lymphopenia can induce compensatory mechanisms to maintain immune homeostasis.
Collapse
Affiliation(s)
- Maryam Nakhaei-Nejad
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Canada
| | - David Barilla
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Chieh-Hsin Lee
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Gregg Blevins
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Fabrizio Giuliani
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Canada
| |
Collapse
|
78
|
Ernst FR, Barr P, Elmor R, Wong SL. Relapse outcomes, safety, and treatment patterns in patients diagnosed with relapsing-remitting multiple sclerosis and initiated on subcutaneous interferon β-1a or dimethyl fumarate: a real-world study. Curr Med Res Opin 2017; 33:2099-2106. [PMID: 28906152 DOI: 10.1080/03007995.2017.1380616] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To estimate real-world treatment patterns, safety, and relapse outcomes of subcutaneous (sc) interferon (IFN) β-1a (Rebif) vs dimethyl fumarate (DMF; Tecfidera), to treat relapsing-remitting multiple sclerosis (RRMS). METHODS A US retrospective chart review of 450 randomly selected adults newly diagnosed with RRMS who received sc IFN β-1a (n = 143) or DMF (n = 307) was conducted. Patients were either (a) treatment-naïve, initiating first-line treatment with sc IFN β-1a or DMF, or (b) previously treated, switching to sc IFN β-1a or DMF. Two years' follow-up data were captured. Patient characteristics, persistence, and adverse events between treatment groups were compared using t-tests or Chi-square tests. Kaplan-Meier curves with log-rank tests and Cox proportional hazards models were used to compare time to, and risk of non-persistence. Annualized Relapse Rates (ARR) were calculated using a robust variance Poisson model adjusting for covariates. Propensity scores were used to address possible selection bias. RESULTS One hundred and twelve patients became non-persistent, most commonly due to an adverse event (n = 37). No difference was observed in time to overall non-persistence between sc IFN β-1a and DMF patients. Among treatment-naïve patients, those receiving DMF had 2.4-times the risk (HR = 2.439, 95% CI = 1.007-5.917, p = .0483) of experiencing a discontinuation than patients receiving sc IFN β-1a. Non-persistent patients receiving DMF had 2.3-times the risk (HR = 2.311, 95% CI = 1.350-3.958, p = .0023) of experiencing an adverse event at a given time point than patients prescribed sc IFN β-1a. No differences in relapse risk or ARR between sc IFN β-1a- and DMF-treated patients were observed. CONCLUSIONS sc IFN β-1a-treated patients had comparable persistence and relapse outcomes, and better safety outcomes vs DMF-treated patients across 2 years.
Collapse
Affiliation(s)
- Frank R Ernst
- a Health Economics and Outcomes Research , Indegene, Inc. , Kennesaw , GA , USA
| | - Peri Barr
- a Health Economics and Outcomes Research , Indegene, Inc. , Kennesaw , GA , USA
| | - Riad Elmor
- a Health Economics and Outcomes Research , Indegene, Inc. , Kennesaw , GA , USA
| | - Schiffon L Wong
- b Global Evidence & Value Development, Global Research & Development , EMD Serono, Inc. , Billerica , MA , USA
| |
Collapse
|
79
|
Calabrese M, Pitteri M, Farina G, Bajrami A, Castellaro M, Magliozzi R, Monaco S. Dimethyl fumarate: a possible exit strategy from natalizumab treatment in patients with multiple sclerosis at risk for severe adverse events. J Neurol Neurosurg Psychiatry 2017; 88:1073-1078. [PMID: 28844068 DOI: 10.1136/jnnp-2017-316236] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/08/2017] [Accepted: 08/06/2017] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Among disease-modifying treatments for multiple sclerosis, natalizumab (NTZ) is highly effective, well tolerated and generally safe. Major concerns regard the risk of developing progressive multifocal leukoencephalopathy (PML), and the occurrence of rebounds or disease activity after its discontinuation. The aim of this study was to explore the efficacy of dimethyl fumarate (DMF) in preventing disease reactivation after NTZ discontinuation. METHODS Thirty-nine patients with relapsing remitting multiple sclerosis, at high risk of PML, were switched from NTZ to DMF and underwent neurological and 3T MRI monitoring for 2 years. Clinical and MRI data regarding the 2-year period preceding NTZ treatment, the 2 years of NTZ treatment and the 2 years of DMF were collected. RESULTS During the DMF phase, among the 39 patients, one or more relapses occurred in five patients (12.8%), increased disability progression in 4 (10.3%) and MRI activity in 8 (20.5%). Post-NTZ rebound effect was observed only in one patient. Overall, only two dropouts (one rebound activity and one gastrointestinal side effect) were registered and almost 80% of the patients have still no evidence of disease activity at the end of DMF treatment. The multiple linear regression model revealed that the number of relapses and MRI parameters before DMF treatment were good predictors of disease activity during treatment with DMF. DISCUSSION DMF appeared generally safe and no carryover PML among investigated cases was observed. Although DMF did not eliminate the possibility of disease reactivation, it seems anyway a promising drug for those patients who shall discontinue NTZ. The clinical and radiological activity preceding the DMF treatment might be used as a prognostic marker of therapy response.
Collapse
Affiliation(s)
- Massimiliano Calabrese
- Neurology B, Department of Neurosciences, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Marco Pitteri
- Neurology B, Department of Neurosciences, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Gabriele Farina
- Neurology B, Department of Neurosciences, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Albulena Bajrami
- Neurology B, Department of Neurosciences, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Marco Castellaro
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Roberta Magliozzi
- Neurology B, Department of Neurosciences, Biomedicine and Movement, University of Verona, Verona, Italy.,Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Salvatore Monaco
- Neurology B, Department of Neurosciences, Biomedicine and Movement, University of Verona, Verona, Italy
| |
Collapse
|
80
|
Han J, Ma S, Gong H, Liu S, Lei L, Hu B, Xu Y, Liu H, Wu D. Inhibition of Acute Graft-versus-Host Disease with Retention of Graft-versus-Tumor Effects by Dimethyl Fumarate. Front Immunol 2017; 8:1605. [PMID: 29209333 PMCID: PMC5702003 DOI: 10.3389/fimmu.2017.01605] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/07/2017] [Indexed: 12/13/2022] Open
Abstract
Acute graft-versus-host disease (aGVHD) remains a clinical challenge and a major source of morbidity and mortality following allogeneic hematopoietic stem cell transplantation (allo-HSCT). Dimethyl fumarate (DMF), an activator of Nrf2, has been shown to have anti-inflammatory and immunomodulatory properties without significant immunosuppression. We therefore hypothesized that DMF could be potentially harnessed for the treatment of aGVHD with retention of graft-versus-tumor effect. In this study, we showed that DMF significantly inhibited alloreactive T cell responses in vitro in mixed lymphocyte reaction assay. Administration of DMF significantly alleviated the severity, histological damage, and the overall mortality of aGVHD in an MHC-mismatched aGVHD model. DMF administration reduced the activation and effector function of donor T cells in vitro and in vivo. In addition, DMF treatment upregulated antioxidant enzymes heme oxygenase-1 and glutathione S-transferase-α1 expressions. Furthermore, DMF treatment markedly increased the frequencies of Treg cells. Depletion of CD25+ cells in DMF recipients aggravated aGVHD mortality compared with IgG control recipients. DMF could promote Treg cell differentiation in a dose dependent manner by upregulating TGF-β expression in vitro. Most importantly, DMF administration preserved graft-versus-leukemia effect after bone marrow transplantation. In conclusion, our findings demonstrated DMF as a promising agent for the prevention of aGVHD after allo-HSCT.
Collapse
Affiliation(s)
- Jingjing Han
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Shoubao Ma
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Huanle Gong
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China
| | - Shuangzhu Liu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Lei Lei
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Bo Hu
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yang Xu
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Haiyan Liu
- Immunology Programme, Department of Microbiology and Immunology, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Depei Wu
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
81
|
Role of Immunological Memory Cells as a Therapeutic Target in Multiple Sclerosis. Brain Sci 2017; 7:brainsci7110148. [PMID: 29112130 PMCID: PMC5704155 DOI: 10.3390/brainsci7110148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/31/2017] [Accepted: 11/02/2017] [Indexed: 12/14/2022] Open
Abstract
Pharmacological targeting of memory cells is an attractive treatment strategy in various autoimmune diseases, such as psoriasis and rheumatoid arthritis. Multiple sclerosis is the most common inflammatory disorder of the central nervous system, characterized by focal immune cell infiltration, activation of microglia and astrocytes, along with progressive damage to myelin sheaths, axons, and neurons. The current review begins with the identification of memory cell types in the previous literature and a recent description of the modulation of these cell types in T, B, and resident memory cells in the presence of different clinically approved multiple sclerosis drugs. Overall, this review paper tries to determine the potential of memory cells to act as a target for the current or newly-developed drugs.
Collapse
|
82
|
Mazzola MA, Raheja R, Regev K, Beynon V, von Glehn F, Paul A, Pierre I, Kivisakk P, Weiner HL, Gandhi R. Monomethyl fumarate treatment impairs maturation of human myeloid dendritic cells and their ability to activate T cells. Mult Scler 2017; 25:63-71. [PMID: 29106333 DOI: 10.1177/1352458517740213] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Dimethyl fumarate (DMF) and its active metabolite monomethyl fumarate (MMF) effectively lead to reduction in disease relapses and active magnetic resonance imaging (MRI) lesions. DMF and MMF are known to be effective in modulating T- and B-cell responses; however, their effect on the phenotype and function of human myeloid dendritic cells (mDCs) is not fully understood. OBJECTIVE To investigate the role of MMF on human mDCs maturation and function. METHODS mDCs from healthy controls were isolated and cultured in vitro with MMF. The effect of MMF on mDC gene expression was determined by polymerase chain reaction (PCR) array after in vitro MMF treatment. The ability of mDCs to activate T cells was assessed by in vitro co-culture system. mDCs from DMF-treated multiple sclerosis (MS) patients were analyzed by flow cytometry and PCR. RESULTS MMF treatment induced a less mature phenotype of mDCs with reduced expression of major histocompatibility complex class II (MHC-II), co-stimulatory molecules CD86, CD40, CD83, and expression of nuclear factor κB (NF-κB) subunits RELA and RELB. mDCs from DMF-treated MS patients also showed the same immature phenotype. T cells co-cultured with MMF-treated mDCs showed reduced proliferation with decreased production of interferon gamma (IFN-γ), interleukin-17 (IL-17), and granulocyte-macrophage colony-stimulating factor (GM-CSF) compared to untreated cells. CONCLUSION We report that MMF can modulate immune response by affecting human mDC function.
Collapse
Affiliation(s)
- Maria Antonietta Mazzola
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Radhika Raheja
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Keren Regev
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vanessa Beynon
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA/Partners MS Center, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Felipe von Glehn
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anu Paul
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Isabelle Pierre
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pia Kivisakk
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA/Partners MS Center, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Roopali Gandhi
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
83
|
Auricchio F, Scavone C, Cimmaruta D, Di Mauro G, Capuano A, Sportiello L, Rafaniello C. Drugs approved for the treatment of multiple sclerosis: review of their safety profile. Expert Opin Drug Saf 2017; 16:1359-1371. [PMID: 28976217 DOI: 10.1080/14740338.2017.1388371] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic immune-mediated inflammatory disorder of the brain and spinal cord characterized by inflammation, demyelination, and axonal degeneration. Area covered: Even though the pharmacological armamentarium for MS treatment is considerably improved in the last 20 years, safety data especially for the second-line and innovative treatments are lacking. In order to analyze the safety profile of drugs used for the treatment of MS, a literature review of pre-marketing, post-marketing studies and case reports was performed. Expert opinion: Nowadays, the numerous drugs approved in the last years for the treatment of MS allow a better control of the disease and a better patient compliance. The main advantages of the new disease-modifying agents for MS (DMTs), in fact, derive from the new oral administration and the prolonged half-life with consequent improvement in compliance compared to first-line therapy which required subcutaneous administrations. However, DMTs can cause serious, sometimes life-threatening or fatal, drug adverse reactions. Due to the lack of safety data and given the recent marketing approval of the last DMTs for MS, observational studies and post-marketing surveillance activities will be necessary in order to improve the knowledge about the safety profile of these drugs and the improvement of their use in clinical practice.
Collapse
Affiliation(s)
- Fabiana Auricchio
- a Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", School of Medicine , University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Cristina Scavone
- a Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", School of Medicine , University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Daniela Cimmaruta
- a Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", School of Medicine , University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Gabriella Di Mauro
- a Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", School of Medicine , University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Annalisa Capuano
- a Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", School of Medicine , University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Liberata Sportiello
- a Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", School of Medicine , University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Concetta Rafaniello
- a Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", School of Medicine , University of Campania "Luigi Vanvitelli" , Naples , Italy
| |
Collapse
|
84
|
Breuer J, Herich S, Schneider-Hohendorf T, Chasan AI, Wettschureck N, Gross CC, Loser K, Zarbock A, Roth J, Klotz L, Wiendl H, Schwab N. Dual action by fumaric acid esters synergistically reduces adhesion to human endothelium. Mult Scler 2017; 24:1871-1882. [PMID: 28984166 DOI: 10.1177/1352458517735189] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Dimethyl fumarate (DMF) is prescribed against relapsing-remitting multiple sclerosis (MS). Here, we investigated the effects of DMF and monomethyl fumarate (MMF), its metabolite in vivo, at the (inflamed) blood-brain barrier (BBB). METHODS Effects of fumaric acid esters were analyzed using primary human brain-derived microvascular endothelial cells (HBMECs) in combination with peripheral blood mononuclear cells (PBMCs) derived from DMF-treated MS patients. RESULTS MMF-binding to brain endothelium cells leads to activation of nuclear factor (erythroid-derived 2)-related factor 2 (Nrf2)-induced downregulation of vascular cell adhesion molecule 1 (VCAM-1). This might be mediated via the G-protein-coupled receptor (GPCR) hydroxycarboxylic acid receptor 2 (HCA2), a known molecular target of MMF, as we could demonstrate its expression and regulation on HBMECs. DMF treatment in vivo led to a strongly reduced expression of VCAM-1's ligand very late antigen 4 (VLA-4) by selectively reducing integrin high-expressing memory T cells of MS patients, potentially due to inhibition of their maturation by reduced trans-localization of NFκB. CONCLUSION DMF-mediated VCAM-1 downregulation on the endothelial side and reduction in T cells with a migratory phenotype on the lymphocyte side result in a synergistic reduction in T-cell adhesion to activated endothelium and, therefore, to reduced BBB transmigration in the setting of MS.
Collapse
Affiliation(s)
- Johanna Breuer
- Department of Neurology, University of Münster, Münster, Germany
| | - Sebastian Herich
- Department of Neurology, University of Münster, Münster, Germany
| | | | - Achmet I Chasan
- Institute of Immunology, University of Münster, Münster, Germany
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany/Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | | | - Karin Loser
- Department of Dermatology, University of Münster, Münster, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, University of Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Luisa Klotz
- Department of Neurology, University of Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Münster, Germany
| | - Nicholas Schwab
- Department of Neurology, University of Münster, Münster, Germany
| |
Collapse
|
85
|
Diebold M, Sievers C, Bantug G, Sanderson N, Kappos L, Kuhle J, Lindberg RLP, Derfuss T. Dimethyl fumarate influences innate and adaptive immunity in multiple sclerosis. J Autoimmun 2017; 86:39-50. [PMID: 28958667 DOI: 10.1016/j.jaut.2017.09.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 09/18/2017] [Accepted: 09/19/2017] [Indexed: 10/18/2022]
Abstract
INTRODUCTION The mode of action of dimethyl fumarate (DMF), an immunomodulatory treatment for relapsing-remitting multiple sclerosis (RRMS), has not yet been fully elucidated. While in-vitro experiments and animal studies suggest effects on immune cell survival, proliferation, migration and oxidative stress response, corresponding observations from human studies are lacking. This study aims to characterize ex-vivo and in-vivo effects in a cohort of DMF treated RRMS patients. METHODS Blood samples were collected from twenty well-characterized RRMS patients at baseline and after 3, 6 and 12 months of DMF treatment and an age- and gender-matched cohort of 20 healthy individuals at 0 and 3 months. Leukocyte subpopulations, immunoglobulin levels and cytokine secretion were measured. T cells were assessed for their levels of reactive oxygen species (ROS), metabolic status and their proliferative capacity. Levels of antioxidants were determined in serum by mass spectrometry. Responses of monocyte activation markers as well as NFkB and MAPK pathways to DMF were analysed. RESULTS Upon DMF treatment, all lymphocyte subpopulations dropped significantly over the course of 12 months with cytotoxic and effector T cells being affected most significantly. DMF induced cell death and inhibited proliferation of T cells in-vitro. Interestingly, this anti-proliferative effect decreased under treatment. In-vivo DMF treatment led to decreased T cell glycolysis and higher turn-over of antioxidants. In line with these results a significant increase of cytosolic ROS levels after 3 months treatment was detected in T cells. In-vitro DMF treatment reduced NFkB (p65) translocation to the nucleus and MAPK (p38) levels decreased upon stimulation with monomethyl fumarate (MMF) in-vitro and ex-vivo. Consequently, the expression of co-stimulatory molecules like CD40 and CD150 was decreased in antigen presenting cells both in-vitro and ex-vivo. CONCLUSION This study translates knowledge from in-vitro and animal studies on DMF into the clinical setting. Our data suggest that DMF not only alters lymphocyte composition, but also has profound effects on proliferation and induces oxidative stress in T cells. It also acts on innate immunity by reducing the activation status of antigen presenting cells (APCs) via NFkB and MAPK inactivation.
Collapse
Affiliation(s)
- Martin Diebold
- Laboratory of Clinical Neuroimmunology, Neurologic Clinic and Policlinic, Departments of Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Claudia Sievers
- Laboratory of Clinical Neuroimmunology, Neurologic Clinic and Policlinic, Departments of Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Glenn Bantug
- Laboratory of Immunobiology, Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Nicholas Sanderson
- Laboratory of Clinical Neuroimmunology, Neurologic Clinic and Policlinic, Departments of Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Ludwig Kappos
- Laboratory of Clinical Neuroimmunology, Neurologic Clinic and Policlinic, Departments of Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Jens Kuhle
- Laboratory of Clinical Neuroimmunology, Neurologic Clinic and Policlinic, Departments of Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Raija L P Lindberg
- Laboratory of Clinical Neuroimmunology, Neurologic Clinic and Policlinic, Departments of Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Tobias Derfuss
- Laboratory of Clinical Neuroimmunology, Neurologic Clinic and Policlinic, Departments of Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland.
| |
Collapse
|
86
|
Gordon-Lipkin E, Banwell B. An update on multiple sclerosis in children: diagnosis, therapies, and prospects for the future. Expert Rev Clin Immunol 2017; 13:975-989. [PMID: 28738749 DOI: 10.1080/1744666x.2017.1360135] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS), a chronic demyelinating disease of the central nervous system, is increasingly being recognized in children and adolescents. Pediatric MS follows a relapsing-remitting course at onset, with a risk for early cognitive impairment. Areas covered: In this review, we discuss the clinical features of acute demyelinating syndromes in children and risk factors that increase the likelihood of a diagnosis of MS. We also address the application of diagnostic criteria for MS in children, immunological features, therapeutic options and psychosocial considerations for children and adolescents with MS. Expert commentary: Collaborative multicenter clinical trials and research efforts are key to the advancement in understanding the pathophysiology and therapeutic strategies for multiple sclerosis across the lifespan.
Collapse
Affiliation(s)
- Eliza Gordon-Lipkin
- a Department of Neurology and Developmental Medicine , Kennedy Krieger Institute and Johns Hopkins School of Medicine , Baltimore , MD , USA
| | - Brenda Banwell
- b Children's Hospital of Philadelphia , Perelman School of Medicine, University of Pennsylvania , Philadelphia , PA , USA
| |
Collapse
|
87
|
Medina S, Villarrubia N, Sainz de la Maza S, Lifante J, Costa-Frossard L, Roldán E, Picón C, Álvarez-Cermeño JC, Villar LM. Optimal response to dimethyl fumarate associates in MS with a shift from an inflammatory to a tolerogenic blood cell profile. Mult Scler 2017; 24:1317-1327. [PMID: 28653862 DOI: 10.1177/1352458517717088] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND The precise mechanism of action of dimethyl fumarate (DMF) treatment in MS remains unknown. OBJECTIVE To identify the changes in the blood lymphocyte profile of MS patients predicting no evidence of disease activity (NEDA) status after DMF treatment. METHODS We studied blood lymphocyte subsets of 64 MS patients treated with DMF at baseline and after 6 months of treatment by flow cytometry. NEDA (41 patients) or ongoing disease activity (ODA, 23 patients) were monitored after a year of follow-up. RESULTS During treatment, all patients experienced an increase in the naive T cells and a decrease in effector memory ones. However, only NEDA patients showed a significant reduction in central memory CD4+ and CD8+ T cells, memory B cells, CD4+ T cells producing interferon (IFN)-gamma, CD8+ T cells producing tumor necrosis factor-alpha (TNF-alpha), and IFN-gamma and B cells producing TNF-alpha. Additionally, they had an increase in regulatory CD56bright cells not observed in ODA group. After treatment, there was a negative correlation between CD56bright cells and CD8+ T cells producing IFN-gamma and TNF-alpha. CONCLUSION A pro-tolerogenic shift in the blood leukocyte profile associates with an optimal response to DMF in MS.
Collapse
Affiliation(s)
- Silvia Medina
- Servicio de Inmunología, Hospital Universitario Ramón y Cajal, Madrid, Spain; IRYCIS, Madrid, Spain; REEM, Barcelona, Spain
| | - Noelia Villarrubia
- Servicio de Inmunología, Hospital Universitario Ramón y Cajal, Madrid, Spain; IRYCIS, Madrid, Spain; REEM, Barcelona, Spain
| | - Susana Sainz de la Maza
- Servicio de Neurología, Hospital Universitario Ramón y Cajal, Madrid, Spain; IRYCIS, Madrid, Spain; REEM, Barcelona, Spain
| | - José Lifante
- Servicio de Inmunología, Hospital Universitario Ramón y Cajal, Madrid, Spain; IRYCIS, Madrid, Spain; REEM, Barcelona, Spain
| | - Lucienne Costa-Frossard
- Servicio de Neurología, Hospital Universitario Ramón y Cajal, Madrid, Spain; IRYCIS, Madrid, Spain; REEM, Barcelona, Spain
| | - Ernesto Roldán
- Servicio de Inmunología, Hospital Universitario Ramón y Cajal, Madrid, Spain; IRYCIS, Madrid, Spain; REEM, Barcelona, Spain
| | - Carmen Picón
- Servicio de Inmunología, Hospital Universitario Ramón y Cajal, Madrid, Spain; IRYCIS, Madrid, Spain; REEM, Barcelona, Spain
| | - José C Álvarez-Cermeño
- Servicio de Neurologia, Hospital Universitario Ramon y Cajal, Madrid, Spain; IRYCIS, Madrid, Spain; REEM, Barcelona, Spain; Department of Medicine, University of Alcala, Madrid, Spain
| | - Luisa M Villar
- Servicio de Inmunología, Hospital Universitario Ramón y Cajal, Madrid, Spain; IRYCIS, Madrid, Spain; REEM, Barcelona, Spain
| |
Collapse
|
88
|
Longbrake EE, Cantoni C, Chahin S, Cignarella F, Cross AH, Piccio L. Dimethyl fumarate induces changes in B- and T-lymphocyte function independent of the effects on absolute lymphocyte count. Mult Scler 2017; 24:728-738. [PMID: 28480794 DOI: 10.1177/1352458517707069] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Dimethyl fumarate (DMF) is used to treat relapsing multiple sclerosis and causes lymphopenia in a subpopulation of treated individuals. Much remains to be learned about how the drug affects B- and T-lymphocytes. OBJECTIVES To characterize changes in B- and T-cell phenotype and function induced by DMF and to investigate whether low absolute lymphocyte count (ALC) is associated with unique functional changes. METHODS Peripheral blood mononuclear cells (PBMCs) were collected from DMF-treated patients, untreated patients, and healthy controls. A subset of DMF-treated patients was lymphopenic (ALC < 800). Multiparametric flow cytometry was used to evaluate cellular phenotypes. Functional response to non-specific and viral peptide stimulation was assessed. RESULTS DMF reduced circulating memory B-cells regardless of ALC. Follicular T-helper cells (CD4+ CXCR5+) and mucosal invariant T-cells (CD8+ CD161+) were also reduced. DMF reduced T-cell production of pro-inflammatory cytokines in response to polyclonal (PMA/ionomycin) and viral peptide stimulation, regardless of ALC. No differences in activation-induced cell death or circulating progenitors were observed between lymphopenic and non-lymphopenic DMF-treated patients. CONCLUSION These data implicate DMF-induced changes in lymphocytes as an important component of the drug's efficacy and expand our understanding of the functional significance of DMF-induced lymphopenia.
Collapse
Affiliation(s)
| | - Claudia Cantoni
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Salim Chahin
- Department of Neurology, Washington University, St. Louis, MO, USA
| | | | - Anne H Cross
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Laura Piccio
- Department of Neurology, Washington University, St. Louis, MO, USA
| |
Collapse
|
89
|
Fleischer V, Friedrich M, Rezk A, Bühler U, Witsch E, Uphaus T, Bittner S, Groppa S, Tackenberg B, Bar-Or A, Zipp F, Luessi F. Treatment response to dimethyl fumarate is characterized by disproportionate CD8+ T cell reduction in MS. Mult Scler 2017; 24:632-641. [PMID: 28436295 DOI: 10.1177/1352458517703799] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND The effect of dimethyl fumarate (DMF) on circulating lymphocyte subsets and their contribution as predictors of clinical efficacy have not yet been investigated in multiple sclerosis (MS). OBJECTIVE To evaluate lymphocytes and lymphocyte subsets (analyzed 6 months after DMF start) in MS patients with and without disease activity after 1 year of treatment in a retrospective study. METHODS Peripheral blood lymphocyte subsets were analyzed by flow cytometry. Untreated MS patients ( n = 40) were compared to those 6 months after onset of DMF treatment ( n = 51). Clinical and magnetic resonance imaging (MRI) disease activity of DMF-treated patients were assessed in the first year under treatment. RESULTS Stable patients showed significantly lower lymphocytes, CD4+ and CD8+ T cells as well as CD19+ B cells compared to active patients under DMF treatment. Furthermore, an increased CD4/CD8 ratio ( p < 0.025) in stable patients indicated a disproportionate reduction of CD8+ T cells relative to CD4+ T cells. Reduced lymphocytes, CD8+ T cells, and CD19+ B cells 6 months after DMF start allowed prediction of the treatment response in the first year. CONCLUSION DMF treatment response is reflected by lower circulating lymphocytes and specific lymphocyte subsets. Changes in the cellular immune profiles under DMF treatment are clinically relevant and might serve as a surrogate marker of treatment response.
Collapse
Affiliation(s)
- Vinzenz Fleischer
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunology (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michaela Friedrich
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunology (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ayman Rezk
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Ulrike Bühler
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunology (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Esther Witsch
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunology (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Timo Uphaus
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunology (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunology (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sergiu Groppa
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunology (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Björn Tackenberg
- Clinical Neuroimmunology Group, Department of Neurology, Philipps University, Marburg, Germany
| | - Amit Bar-Or
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunology (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Felix Luessi
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunology (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
90
|
Affiliation(s)
- Josep Dalmau
- ICREA-IDIBAPS, Hospital Clínic, University of Barcelona, Spain; and Department of Neurology, University of Pennsylvania
| |
Collapse
|
91
|
Kira JI. Unexpected exacerbations following initiation of disease-modifying drugs in neuromyelitis optica spectrum disorder: Which factor is responsible, anti-aquaporin 4 antibodies, B cells, Th1 cells, Th2 cells, Th17 cells, or others? Mult Scler 2017; 23:1300-1302. [DOI: 10.1177/1352458517703803] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Some disease-modifying drugs for multiple sclerosis, which mainly act on T cells, are ineffective for neuromyelitis optica spectrum disorder and induce unexpected relapses. These include interferon beta, glatiramer acetate, fingolimod, natalizumab, and alemtuzumab. The cases reported here suggest that dimethyl fumarate, which reduces the number of Th1 and Th17 cells and induces IL-4-producing Th2 cells, is also unsuitable for neuromyelitis optica spectrum disorder, irrespective of anti-aquaporin 4 IgG serostatus. Although oral dimethyl fumarate with manageable adverse effects is easy to initiate in the early course of multiple sclerosis, special attention should be paid for atypical demyelinating cases.
Collapse
Affiliation(s)
- Jun-ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
92
|
Chaves C, Ganguly R, Ceresia C, Camac A. Lymphocyte subtypes in relapsing-remitting multiple sclerosis patients treated with dimethyl fumarate. Mult Scler J Exp Transl Clin 2017; 3:2055217317702933. [PMID: 28607757 PMCID: PMC5408504 DOI: 10.1177/2055217317702933] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/12/2017] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Recent data suggest that lymphopenia is more prevalent than reported in relapsing-remitting multiple sclerosis (RRMS) patients taking dimethyl fumarate (DMF). OBJECTIVE The objective of this study was to investigate the effect of DMF on lymphocyte subtypes in RRMS patients with and without lymphopenia. METHOD A retrospective study compared lymphocyte subtypes in DMF-treated RRMS patients with low (G1, n = 35) and normal lymphocyte counts (G2, n = 24). RESULTS Fifty-nine patients were identified, with mean age 49, 71.2% females, and average DMF duration 20 months. Age, sex, baseline white blood count, disease and treatment durations were similar between groups. Prior interferon therapy and baseline lower normal lymphocyte counts were more frequent in G1. Mean lymphocyte counts were 0.8 ± 0.2 × 109/L in G1 and 1.6 ± 0.3 × 109/L in G2. CD3+, CD4+, and CD8+ T cell mean counts were lower (p < 0.0001), while CD4/CD8 ratio higher (p = 0.03) in G1 than G2. Mean CD19 + B cell counts were normal; however, values were lower in G1 (p = 0.04). After adjusting for confounders, significantly positive correlations were noted between lymphocyte counts and CD3 + , CD4+, CD8+ T, and B cell counts. Negative correlation was observed between lymphocyte counts and CD4/CD8 ratio driven by low CD8+ T cell counts. CONCLUSION DMF treatment predominantly impacts T cells, in particular CD8+ subtype. This finding may have implications in this population's immunocompetence.
Collapse
Affiliation(s)
| | | | | | - A Camac
- Neurology Department, Lahey Clinic, Lexington, MA, USA
| |
Collapse
|
93
|
Ghadiri M, Rezk A, Li R, Evans A, Luessi F, Zipp F, Giacomini PS, Antel J, Bar-Or A. Dimethyl fumarate-induced lymphopenia in MS due to differential T-cell subset apoptosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2017; 4:e340. [PMID: 28377940 PMCID: PMC5365096 DOI: 10.1212/nxi.0000000000000340] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 01/30/2017] [Indexed: 01/12/2023]
Abstract
Objective: To examine the mechanism underlying the preferential CD8+ vs CD4+ T-cell lymphopenia induced by dimethyl fumarate (DMF) treatment of MS. Methods: Total lymphocyte counts and comprehensive T-cell subset analyses were performed in high-quality samples obtained from patients with MS prior to and serially following DMF treatment initiation. Random coefficient mixed-effects analysis was used to model the trajectory of T-cell subset losses in vivo. Survival and apoptosis of distinct T-cell subsets were assessed following in vitro exposure to DMF. Results: Best-fit modeling indicated that the DMF-induced preferential reductions in CD8+ vs CD4+ T-cell counts nonetheless followed similar depletion kinetics, suggesting a similar rather than distinct mechanism involved in losses of both the CD8+ and CD4+ T cells. In vitro, DMF exposure resulted in dose-dependent reductions in T-cell survival, which were found to reflect apoptotic cell death. This DMF-induced apoptosis was greater for CD8+ vs CD4+, as well as for memory vs naive, and conventional vs regulatory T-cell subsets, a pattern which mirrored preferential T-cell subset losses that we observed during in vivo treatment of patients. Conclusions: Differential apoptosis mediated by DMF may underlie the preferential lymphopenia of distinct T-cell subsets, including CD8+ and memory T-cell subsets, seen in treated patients with MS. This differential susceptibility of distinct T-cell subsets to DMF-induced apoptosis may contribute to both the safety and efficacy profiles of DMF in patients with MS.
Collapse
Affiliation(s)
- Mahtab Ghadiri
- Montreal Neurological Institute (M.G., A.R., R.L., P.S.G., J.A., A.B.-O.), McGill University, Montreal, QC, Canada; Brain and Mind Centre (M.G.), University of Sydney, NSW, Australia; Institute of Actuaries of Australia (A.E.); Department of Neurology (F.L., F.Z.), University Medical Center Mainz, Germany; and Department of Neurology (A.R., R.L., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Ayman Rezk
- Montreal Neurological Institute (M.G., A.R., R.L., P.S.G., J.A., A.B.-O.), McGill University, Montreal, QC, Canada; Brain and Mind Centre (M.G.), University of Sydney, NSW, Australia; Institute of Actuaries of Australia (A.E.); Department of Neurology (F.L., F.Z.), University Medical Center Mainz, Germany; and Department of Neurology (A.R., R.L., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Rui Li
- Montreal Neurological Institute (M.G., A.R., R.L., P.S.G., J.A., A.B.-O.), McGill University, Montreal, QC, Canada; Brain and Mind Centre (M.G.), University of Sydney, NSW, Australia; Institute of Actuaries of Australia (A.E.); Department of Neurology (F.L., F.Z.), University Medical Center Mainz, Germany; and Department of Neurology (A.R., R.L., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Ashley Evans
- Montreal Neurological Institute (M.G., A.R., R.L., P.S.G., J.A., A.B.-O.), McGill University, Montreal, QC, Canada; Brain and Mind Centre (M.G.), University of Sydney, NSW, Australia; Institute of Actuaries of Australia (A.E.); Department of Neurology (F.L., F.Z.), University Medical Center Mainz, Germany; and Department of Neurology (A.R., R.L., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Felix Luessi
- Montreal Neurological Institute (M.G., A.R., R.L., P.S.G., J.A., A.B.-O.), McGill University, Montreal, QC, Canada; Brain and Mind Centre (M.G.), University of Sydney, NSW, Australia; Institute of Actuaries of Australia (A.E.); Department of Neurology (F.L., F.Z.), University Medical Center Mainz, Germany; and Department of Neurology (A.R., R.L., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Frauke Zipp
- Montreal Neurological Institute (M.G., A.R., R.L., P.S.G., J.A., A.B.-O.), McGill University, Montreal, QC, Canada; Brain and Mind Centre (M.G.), University of Sydney, NSW, Australia; Institute of Actuaries of Australia (A.E.); Department of Neurology (F.L., F.Z.), University Medical Center Mainz, Germany; and Department of Neurology (A.R., R.L., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Paul S Giacomini
- Montreal Neurological Institute (M.G., A.R., R.L., P.S.G., J.A., A.B.-O.), McGill University, Montreal, QC, Canada; Brain and Mind Centre (M.G.), University of Sydney, NSW, Australia; Institute of Actuaries of Australia (A.E.); Department of Neurology (F.L., F.Z.), University Medical Center Mainz, Germany; and Department of Neurology (A.R., R.L., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jack Antel
- Montreal Neurological Institute (M.G., A.R., R.L., P.S.G., J.A., A.B.-O.), McGill University, Montreal, QC, Canada; Brain and Mind Centre (M.G.), University of Sydney, NSW, Australia; Institute of Actuaries of Australia (A.E.); Department of Neurology (F.L., F.Z.), University Medical Center Mainz, Germany; and Department of Neurology (A.R., R.L., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Amit Bar-Or
- Montreal Neurological Institute (M.G., A.R., R.L., P.S.G., J.A., A.B.-O.), McGill University, Montreal, QC, Canada; Brain and Mind Centre (M.G.), University of Sydney, NSW, Australia; Institute of Actuaries of Australia (A.E.); Department of Neurology (F.L., F.Z.), University Medical Center Mainz, Germany; and Department of Neurology (A.R., R.L., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
94
|
Wu Q, Wang Q, Mao G, Dowling CA, Lundy SK, Mao-Draayer Y. Dimethyl Fumarate Selectively Reduces Memory T Cells and Shifts the Balance between Th1/Th17 and Th2 in Multiple Sclerosis Patients. THE JOURNAL OF IMMUNOLOGY 2017; 198:3069-3080. [PMID: 28258191 DOI: 10.4049/jimmunol.1601532] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 02/08/2017] [Indexed: 01/20/2023]
Abstract
Dimethyl fumarate (DMF; trade name Tecfidera) is an oral formulation of the fumaric acid ester that is Food and Drug Administration approved for treatment of relapsing-remitting multiple sclerosis. To better understand the therapeutic effects of Tecfidera and its rare side effect of progressive multifocal leukoencephalopathy, we conducted cross-sectional and longitudinal studies by immunophenotyping cells from peripheral blood (particularly T lymphocytes) derived from untreated and 4-6 and 18-26 mo Tecfidera-treated stable relapsing-remitting multiple sclerosis patients using multiparametric flow cytometry. The absolute numbers of CD4 and CD8 T cells were significantly decreased and the CD4/CD8 ratio was increased with DMF treatment. The proportions of both effector memory T cells and central memory T cells were reduced, whereas naive T cells increased in treated patients. T cell activation was reduced with DMF treatment, especially among effector memory T cells and effector memory RA T cells. Th subsets Th1 (CXCR3+), Th17 (CCR6+), and particularly those expressing both CXCR3 and CD161 were reduced most significantly, whereas the anti-inflammatory Th2 subset (CCR3+) was increased after DMF treatment. A corresponding increase in IL-4 and decrease in IFN-γ and IL-17-expressing CD4+ T cells were observed in DMF-treated patients. DMF in vitro treatment also led to increased T cell apoptosis and decreased activation, proliferation, reactive oxygen species, and CCR7 expression. Our results suggest that DMF acts on specific memory and effector T cell subsets by limiting their survival, proliferation, activation, and cytokine production. Monitoring these subsets could help to evaluate the efficacy and safety of DMF treatment.
Collapse
Affiliation(s)
- Qi Wu
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Qin Wang
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Guangmei Mao
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Catherine A Dowling
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Steven K Lundy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109; and.,Graduate Program in Immunology, Program in Biomedical Sciences, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109; .,Graduate Program in Immunology, Program in Biomedical Sciences, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
95
|
Klotz L, Berthele A, Brück W, Chan A, Flachenecker P, Gold R, Haghikia A, Hellwig K, Hemmer B, Hohlfeld R, Korn T, Kümpfel T, Lang M, Limmroth V, Linker RA, Meier U, Meuth SG, Paul F, Salmen A, Stangel M, Tackenberg B, Tumani H, Warnke C, Weber MS, Ziemssen T, Zipp F, Wiendl H. [Monitoring of blood parameters under course-modified MS therapy : Substance-specific relevance and current recommendations for action]. DER NERVENARZT 2017; 87:645-59. [PMID: 26927677 DOI: 10.1007/s00115-016-0077-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
With the approval of various substances for the immunotherapy of multiple sclerosis (MS), treatment possibilities have improved significantly over the last few years. Indeed, the choice of individually tailored preparations and treatment monitoring for the treating doctor is becoming increasingly more complex. This is particularly applicable for monitoring for a treatment-induced compromise of the immune system. The following article by members of the German Multiple Sclerosis Skills Network (KKNMS) and the task force "Provision Structures and Therapeutics" summarizes the practical recommendations for approved immunotherapy for mild to moderate and for (highly) active courses of MS. The focus is on elucidating the substance-specific relevance of particular laboratory parameters with regard to the mechanism of action and the side effects profile. To enable appropriate action to be taken in clinical practice, any blood work changes that can be expected, in addition to any undesirable laboratory findings and their causes and relevance, should be elucidated.
Collapse
Affiliation(s)
- L Klotz
- Department für Neurologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Deutschland
| | - A Berthele
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar der TU München, Ismaninger Straße 22, 81675, München, Deutschland
| | - W Brück
- Institut für Neuropathologie, Universitätsmedizin Göttingen der Georg-August-Universität, Robert-Koch-Str. 40, 37075, Göttingen, Deutschland
| | - A Chan
- Neurologische Klinik, St. Josef-Hospital, Universitätsklinikum der Ruhr-Universität Bochum, Gudrunstr. 56, 44791, Bochum, Deutschland
| | - P Flachenecker
- Neurologisches Rehabilitationszentrum Quellenhof in Bad Wildbad GmbH, Kuranlagenallee 2, 75323, Bad Wildbad, Deutschland
| | - R Gold
- Neurologische Klinik, St. Josef-Hospital, Universitätsklinikum der Ruhr-Universität Bochum, Gudrunstr. 56, 44791, Bochum, Deutschland
| | - A Haghikia
- Neurologische Klinik, St. Josef-Hospital, Universitätsklinikum der Ruhr-Universität Bochum, Gudrunstr. 56, 44791, Bochum, Deutschland
| | - K Hellwig
- Neurologische Klinik, St. Josef-Hospital, Universitätsklinikum der Ruhr-Universität Bochum, Gudrunstr. 56, 44791, Bochum, Deutschland
| | - B Hemmer
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar der TU München, Ismaninger Straße 22, 81675, München, Deutschland
| | - R Hohlfeld
- Institut für Klinische Neuroimmunologie, Klinikum der Universität München, Campus Großhadern, Marchioninistr. 15, 81377, München, Deutschland
| | - T Korn
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar der TU München, Ismaninger Straße 22, 81675, München, Deutschland
| | - T Kümpfel
- Institut für Klinische Neuroimmunologie, Klinikum der Universität München, Campus Großhadern, Marchioninistr. 15, 81377, München, Deutschland
| | - M Lang
- NeuroTransConcept GmbH, Centers of Excellence, Pfauengasse 8, 89073, Ulm, Deutschland
| | - V Limmroth
- Klinik für Neurologie und Palliativmedizin, Kliniken der Stadt Köln, Ostmerheimer Str. 200, 51109, Köln - Merheim, Deutschland
| | - R A Linker
- Neurologische Klinik, Universitätsklinikum Erlangen, Schwabachanlage 6, 91054, Erlangen, Deutschland
| | - U Meier
- Berufsverband Deutscher Neurologen BDN, Am Ziegelkamp 1f, 41515, Grevenbroich, Deutschland
| | - S G Meuth
- Department für Neurologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Deutschland
| | - F Paul
- Institut für Neuroimmunologie, Universitätsklinikum Charité, Schumannstr. 20/21, 10117, Berlin, Deutschland
| | - A Salmen
- Neurologische Klinik, St. Josef-Hospital, Universitätsklinikum der Ruhr-Universität Bochum, Gudrunstr. 56, 44791, Bochum, Deutschland
| | - M Stangel
- Klinik für Neurologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, 30625, Hannover, Deutschland
| | - B Tackenberg
- Klinik für Neurologie, Philipps-Universität und Universitätsklinikum Marburg, Baldingerstr. 1, 35043, Marburg, Deutschland
| | - H Tumani
- Neurologische Universitätsklinik der Universität Ulm, Oberer Eselsberg 45, 89081, Ulm, Deutschland.,Fachklinik für Neurologie Dietenbronn, Dietenbronn 7, 88477, Schwendi, Deutschland
| | - C Warnke
- Klinik für Neurologie, Universitätsklinikum Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Deutschland
| | - M S Weber
- Institut für Neuropathologie, Universitätsmedizin Göttingen der Georg-August-Universität, Robert-Koch-Str. 40, 37075, Göttingen, Deutschland
| | - T Ziemssen
- Klinik und Poliklinik für Neurologie, Universitätsklinikum Carl Gustav Carus der TU Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland
| | - F Zipp
- Klinik für Neurologie, Universitätsmedizin der Johannes-Gutenberg-Universität Mainz, Langenbeckstr. 1, 55131, Mainz, Deutschland
| | - H Wiendl
- Department für Neurologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Deutschland.
| |
Collapse
|
96
|
Abstract
PURPOSE OF REVIEW Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system most often characterized by clinical relapses and periods of remission. RECENT FINDINGS The past decade has seen a dramatic increase in disease-modifying therapies for MS. Fourteen FDA-approved immunomodulatory drugs are currently available, and more medications are in development. A growing number of reported opportunistic infections, including progressive multifocal leukoencephalopathy (PML), highlight the serious complications of these new drugs and the need for specific screening guidelines. Using data from Phase II and III randomized controlled trials, case reports, drug manufacturing data, and clinical experience, we outline the most common and serious infections associated with novel MS therapies.
Collapse
|
97
|
Schlöder J, Berges C, Luessi F, Jonuleit H. Dimethyl Fumarate Therapy Significantly Improves the Responsiveness of T Cells in Multiple Sclerosis Patients for Immunoregulation by Regulatory T Cells. Int J Mol Sci 2017; 18:ijms18020271. [PMID: 28134847 PMCID: PMC5343807 DOI: 10.3390/ijms18020271] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 01/14/2017] [Accepted: 01/22/2017] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease caused by an insufficient suppression of autoreactive T lymphocytes. One reason for the lack of immunological control is the reduced responsiveness of T effector cells (Teff) for the suppressive properties of regulatory T cells (Treg), a process termed Treg resistance. Here we investigated whether the disease-modifying therapy of relapsing-remitting MS (RRMS) with dimethyl fumarate (DMF) influences the sensitivity of T cells in the peripheral blood of patients towards Treg-mediated suppression. We demonstrated that DMF restores responsiveness of Teff to the suppressive function of Treg in vitro, presumably by down-regulation of interleukin-6R (IL-6R) expression on T cells. Transfer of human immune cells into immunodeficient mice resulted in a lethal graft-versus-host reaction triggered by human CD4⁺ Teff. This systemic inflammation can be prevented by activated Treg after transfer of immune cells from DMF-treated MS patients, but not after injection of Treg-resistant Teff from therapy-naïve MS patients. Furthermore, after DMF therapy, proliferation and expansion of T cells and the immigration into the spleen of the animals is reduced and modulated by activated Treg. In summary, our data reveals that DMF therapy significantly improves the responsiveness of Teff in MS patients to immunoregulation.
Collapse
Affiliation(s)
- Janine Schlöder
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Carsten Berges
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Felix Luessi
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Helmut Jonuleit
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
98
|
Havla J, Warnke C, Derfuss T, Kappos L, Hartung HP, Hohlfeld R. Interdisciplinary Risk Management in the Treatment of Multiple Sclerosis. DEUTSCHES ARZTEBLATT INTERNATIONAL 2016; 113:879-886. [PMID: 28130920 PMCID: PMC5282476 DOI: 10.3238/arztebl.2016.0879] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 06/20/2016] [Accepted: 10/05/2016] [Indexed: 01/14/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is the most common autoimmune disease of the central nervous system. There are at least 150 000 persons with MS in Germany. Recent years have seen the approval of new drugs against. METHODS This article is based on pertinent literature retrieved by a selective search in PubMed as well as on documentation of relevant risks and adverse effects in "red hand letters" (information bulletins from pharmaceutical companies to physicians about adverse drug effects) and elsewhere, along with data provided by the German Multiple Sclerosis Competence Network. RESULTS In recent years, there have been major advances enabling better, more individualized treatment of patients with MS. Physicians must, however, give due consideration to potentially severe or even life-threatening adverse drug effects. These can include, for example, transaminase elevation (hepatotoxicity), cardio- and nephrotoxicity, or lympho- and leukopenia with a variable risk of infection. Among patients taking natalizumab, the cumulative risk of developing progressive multifocal leukencephalopathy (PML) may be 1:100 or higher, depending on the individual risk profile. Rare cases of PML have also been seen under treatment with fingolimod and dimethyl fumarate. Moreover, any type of immunosuppressive treatment can, at least theoretically, increase the risk of malignant disease. Secondary autoimmune diseases can arise as well: approximately 35% of patients treated with alemtuzumab develop autoimmune thyroid disease within two years, and 2% of patients who take daclizumab have severe autoimmune dermatological side effects. Teriflunomide, fingolimod, natalizumab, mitoxantrone, interferon β1-a/b, and daclizumab can all damage the liver. There are also psychiatric, reproductive, and vaccineassociated risks and side effects that must be considered. CONCLUSION Newer drugs for MS have enabled more effective treatment, but are also associated with a higher risk of side effects. Interdisciplinary risk management is needed.
Collapse
Affiliation(s)
- Joachim Havla
- Institute for Clinical Neuroimmunology, Biomedical Center and Hospital, Ludwig-Maximilians Universität München, Munich
| | - Clemens Warnke
- Department of Neurology, Faculty of Medicine, Heinrich Heine University Düsseldorf
| | | | | | - Hans-Peter Hartung
- Department of Neurology, Faculty of Medicine, Heinrich Heine University Düsseldorf
| | - Reinhard Hohlfeld
- Institute for Clinical Neuroimmunology, Biomedical Center and Hospital, Ludwig-Maximilians Universität München, Munich
- Munich Cluster for Systems Neurology (SyNergy)
| |
Collapse
|
99
|
Gross CC, Schulte-Mecklenbeck A, Wiendl H, Marcenaro E, Kerlero de Rosbo N, Uccelli A, Laroni A. Regulatory Functions of Natural Killer Cells in Multiple Sclerosis. Front Immunol 2016; 7:606. [PMID: 28066417 PMCID: PMC5165263 DOI: 10.3389/fimmu.2016.00606] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 12/01/2016] [Indexed: 12/26/2022] Open
Abstract
There is increasing evidence that natural killer (NK) cells exhibit regulatory features. Among them, CD56bright NK cells have been suggested to play a major role in controlling T cell responses and maintaining homeostasis. Dysfunction in NK cell-mediated regulatory features has been recently described in untreated multiple sclerosis (MS), suggesting a contribution to MS pathogenesis. Moreover, biological disease-modifying treatments effective in MS apparently enhance the frequencies and/or regulatory function of NK cells, further pointing toward an immunoprotective role of NK cells in MS. Here, we summarize the current knowledge on the regulatory functions of NK cells, based on their interactions with other cells belonging to the innate compartment, as well as with adaptive effector cells. We review the more recent data reporting disruption of NK cell/T cell interactions in MS and discuss how disease-modifying treatments for MS affect NK cells.
Collapse
Affiliation(s)
- Catharina C Gross
- Department of Neurology, University Hospital Münster , Münster , Germany
| | | | - Heinz Wiendl
- Department of Neurology, University Hospital Münster , Münster , Germany
| | - Emanuela Marcenaro
- Centre of Excellence for Biomedical Research, University of Genova, Genova, Italy; Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Nicole Kerlero de Rosbo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova , Genova , Italy
| | - Antonio Uccelli
- Centre of Excellence for Biomedical Research, University of Genova, Genova, Italy; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy; IRCCS San Martino-IST, Genova, Italy
| | - Alice Laroni
- Centre of Excellence for Biomedical Research, University of Genova, Genova, Italy; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy; IRCCS San Martino-IST, Genova, Italy
| |
Collapse
|
100
|
Li R, Rezk A, Ghadiri M, Luessi F, Zipp F, Li H, Giacomini PS, Antel J, Bar-Or A. Dimethyl Fumarate Treatment Mediates an Anti-Inflammatory Shift in B Cell Subsets of Patients with Multiple Sclerosis. THE JOURNAL OF IMMUNOLOGY 2016; 198:691-698. [PMID: 27974457 DOI: 10.4049/jimmunol.1601649] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/14/2016] [Indexed: 11/19/2022]
Abstract
The therapeutic mode of action of dimethyl fumarate (DMF), approved for treating patients with relapsing-remitting multiple sclerosis, is not fully understood. Recently, we and others demonstrated that Ab-independent functions of distinct B cell subsets are important in mediating multiple sclerosis (MS) relapsing disease activity. Our objective was to test whether and how DMF influences both the phenotype and functional responses of disease-implicated B cell subsets in patients with MS. High-quality PBMC were obtained from relapsing-remitting MS patients prior to and serially after initiation of DMF treatment. Multiparametric flow cytometry was used to monitor the phenotype and functional response-profiles of distinct B cell subsets. Total B cell counts decreased following DMF treatment, largely reflecting losses of circulating mature/differentiated (but not of immature transitional) B cells. Within the mature B cell pool, DMF had a greater impact on memory than naive B cells. In keeping with these in vivo effects, DMF treatment in vitro remarkably diminished mature (but not transitional B cell) survival, mediated by inducing apoptotic cell death. Although DMF treatment (both in vivo and in vitro) minimally impacted B cell IL-10 expression, it strongly reduced B cell expression of GM-CSF, IL-6, and TNF-α, resulting in a significant anti-inflammatory shift of B cell response profiles. The DMF-mediated decrease in B cell proinflammatory cytokine responses was further associated with reduced phosphorylation of STAT5/6 and NF-κB in surviving B cells. Together, these data implicate novel mechanisms by which DMF may modulate MS disease activity through shifting the balance between pro- and anti-inflammatory B cell responses.
Collapse
Affiliation(s)
- Rui Li
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Ayman Rezk
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Mathab Ghadiri
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Felix Luessi
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada.,Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine-Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany; and
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine-Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany; and
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University, NanGang District, Harbin 150086, Heilongjiang, China
| | - Paul S Giacomini
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jack Antel
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Amit Bar-Or
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada;
| |
Collapse
|