151
|
Montalban X, Gold R, Thompson AJ, Otero‐Romero S, Amato MP, Chandraratna D, Clanet M, Comi G, Derfuss T, Fazekas F, Hartung HP, Havrdova E, Hemmer B, Kappos L, Liblau R, Lubetzki C, Marcus E, Miller DH, Olsson T, Pilling S, Selmaj K, Siva A, Sorensen PS, Sormani MP, Thalheim C, Wiendl H, Zipp F. ECTRIMS
/
EAN
guideline on the pharmacological treatment of people with multiple sclerosis. Eur J Neurol 2018; 25:215-237. [DOI: 10.1111/ene.13536] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 11/27/2017] [Indexed: 01/21/2023]
Affiliation(s)
- X. Montalban
- Multiple Sclerosis Centre of Catalonia (Cemcat) Department of Neurology‐Neuroimmunology Vall d'Hebron University Hospital Barcelona Spain
| | - R. Gold
- Department of Neurology Ruhr University, St Josef‐Hospital Bochum Germany
| | - A. J. Thompson
- Department of Brain Repair and Rehabilitation Faculty of Brain Sciences University College London Institute of Neurology London UK
| | - S. Otero‐Romero
- Multiple Sclerosis Centre of Catalonia (Cemcat) Department of Neurology‐Neuroimmunology Vall d'Hebron University Hospital Barcelona Spain
- Preventive Medicine and Epidemiology Department Vall d'Hebron University Hospital Barcelona Spain
| | - M. P. Amato
- Department NEUROFARBA Section Neurosciences University of Florence Florence Italy
| | | | - M. Clanet
- Department of Neurology Toulouse University Hospital Toulouse France
| | - G. Comi
- Neurological Department Institute of Experimental Neurology (INSPE) Scientific Institute Hospital San Raffaele University Vita‐Salute San Raffaele Milan Italy
| | - T. Derfuss
- Departments of Neurology and Biomedicine University Hospital Basel Basel Switzerland
| | - F. Fazekas
- Department of Neurology Medical University of Graz Graz Austria
| | - H. P. Hartung
- Department of Neurology Medical Faculty, Multiple Sclerosis Heinrich‐Heine‐University Düsseldorf Germany
| | - E. Havrdova
- Department of Neurology and Centre of Clinical Neuroscience First Faculty of Medicine and General University Hospital Charles University Prague Czech Republic
| | - B. Hemmer
- Department of Neurology Klinikum Rechts der Isar Technische Universität München MunichGermany
- Munich Cluster for Systems Neurology (SyNergy) Munich Germany
| | - L. Kappos
- University Hospital Basel Basel Switzerland
| | - R. Liblau
- INSERM UMR U1043 – CNRS U5282 Centre de Physiopathologie de Toulouse Purpan Université de Toulouse, UPS ToulouseFrance
| | - C. Lubetzki
- ICM‐GHU Pitié‐Salpêtrière Sorbonne Universités UPMC Univ Paris 06, UMR_S 1127 Paris France
| | - E. Marcus
- Centre for Outcomes Research and Effectiveness (CORE) Research Department of Clinical, Educational and Health Psychology University College London LondonUK
| | - D. H. Miller
- NMR Research Unit Queen Square Multiple Sclerosis Centre University College London (UCL) Institute of Neurology London UK
| | - T. Olsson
- Neuroimmunology Unit Centre for Molecular Medicine, L8:04 Karolinska University Hospital (Solna) Stockholm Sweden
| | - S. Pilling
- Centre for Outcomes Research and Effectiveness (CORE) Research Department of Clinical, Educational and Health Psychology University College London LondonUK
| | - K. Selmaj
- Department of Neurology Medical University of Lodz Lodz Poland
| | - A. Siva
- Clinical Neuroimmunology Unit and MS Clinic Department of Neurology Cerrahpasa School of Medicine Istanbul University Istanbul Turkey
| | - P. S. Sorensen
- Danish Multiple Sclerosis Centre Department of Neurology Copenhagen University Hospital Rigshospitalet Copenhagen Denmark
| | | | - C. Thalheim
- European Multiple Sclerosis Platform (EMSP) Schaerbeek/Brussels Belgium
| | - H. Wiendl
- Department of Neurology University of Münster MünsterGermany
| | - F. Zipp
- Department of Neurology Focus Program Translational Neuroscience (FTN) and Immunology (FZI) Rhine‐Main Neuroscience Network (rmn2) University Medical Centre of the Johannes Gutenberg University Mainz Mainz Germany
| |
Collapse
|
152
|
Miller AE. Switching or Discontinuing Disease-Modifying Therapies for Multiple Sclerosis. Continuum (Minneap Minn) 2018; 22:851-63. [PMID: 27261686 DOI: 10.1212/con.0000000000000327] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE OF REVIEW This article reviews the reasons for discontinuation or switching of multiple sclerosis disease-modifying therapy as well as procedures that might mitigate risk to the patient under such circumstances. RECENT FINDINGS Recent review of the literature, as well as the author's extensive clinical experience, indicate that the discontinuation of multiple sclerosis disease-modifying therapies occurs for many reasons. Often one medication is stopped at the recommendation of the physician in order to switch to another medication. However, often the decision to discontinue medication is made by the patient. Unfortunately, in still other situations, treatment is stopped because of circumstances beyond the control of either patient or physician (eg, a loss of insurance coverage). Currently available data do not permit a conclusion about whether it is ever safe to discontinue disease-modifying therapy in a stable patient without the expectation of return of disease activity. SUMMARY Clinicians must help patients avoid unnecessary and undesirable cessation of disease-modifying therapy. While switches of therapy are often necessary, steps to minimize both adverse events and the risk of recurrent disease should be undertaken. Whether disease-modifying therapy can ever be purposely discontinued without incurring a significant risk of disease recurrence remains to be determined.
Collapse
|
153
|
Abstract
PURPOSE OF REVIEW Many therapeutic advances for relapsing-remitting multiple sclerosis (MS) have occurred in the past 25 years. Although similar advances in disease-modifying therapies have not been realized in progressive MS, many symptomatic therapeutic strategies can benefit patients with progressive MS. Few guidelines exist for management of patients with progressive MS. RECENT FINDINGS The classification of progressive MS was revised in 2013 to include a description of inflammatory disease activity determined by clinical relapses or imaging findings. Developing knowledge about the pathogenesis of progressive MS and the role of comorbidities in modifying the disease course has implications for the clinical management of patients with progressive MS as well as for clinical trial design. Current and upcoming clinical trials will assess a wide range of interventions, including immunomodulatory agents, putative neuroprotective molecules, stem cell therapy, nutrition, and rehabilitation techniques. SUMMARY None of the therapies currently approved for use in relapsing-remitting MS have been shown to slow the gradual progression of disability that occurs in the absence of recent relapses or changes in MRI. A multidisciplinary approach is needed to address the many symptoms that impact quality of life for patients with progressive MS.
Collapse
|
154
|
Rodrigues DB, Oliveira JM, Santos TC, Reis RL. Dendrimers: Breaking the paradigm of current musculoskeletal autoimmune therapies. J Tissue Eng Regen Med 2018; 12:e1796-e1812. [DOI: 10.1002/term.2597] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 09/01/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Daniel B. Rodrigues
- 3B's Research Group – Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineUniversity of Minho Avepark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Joaquim M. Oliveira
- 3B's Research Group – Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineUniversity of Minho Avepark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineHeadquarters at University of Minho Avepark 4805‐017 Barco Guimarães Portugal
| | - Tírcia C. Santos
- 3B's Research Group – Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineUniversity of Minho Avepark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Rui L. Reis
- 3B's Research Group – Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineUniversity of Minho Avepark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineHeadquarters at University of Minho Avepark 4805‐017 Barco Guimarães Portugal
| |
Collapse
|
155
|
Abstract
Accumulating neurological disability has a substantial impact on the lives of patients with multiple sclerosis (MS). As well as the established Expanded Disability Status Scale (EDSS), several other outcome measures are now available for assessing disability progression in MS. This review extends the findings of a previous analysis of relapsing-remitting MS (RRMS) trials published up to 2012, to determine whether there has been a shift in outcome measures used to assess disability in phase III clinical trials in RRMS and progressive MS. Forty relevant trials were identified (RRMS, n = 16; progressive MS, n = 18; other/mixed phenotypes, n = 6). Sustained EDSS worsening, particularly over 3 months, was included as an endpoint in almost all identified trials. Other disability-related endpoints included the Multiple Sclerosis Functional Composite z-score and scores for the physical component summary of the Multiple Sclerosis Impact Scale and Medical Outcomes Study Short-Form (36-item) Health Survey. Tests assessing manual dexterity, ambulation, vision and cognition were also employed, and in some trials, composite endpoints were used. However, there was no obvious trend in choice of disability outcome measures over time. Sustained EDSS worsening over short time periods continues to be the most widely used measure of disability progression in pivotal MS trials, despite its well-recognised limitations. A new tool set is needed for use in MS clinical trials that detects the benefit of potential treatments that slow (or reverse) progressive disability.
Collapse
|
156
|
Adiele RC, Adiele CA. Metabolic defects in multiple sclerosis. Mitochondrion 2017; 44:7-14. [PMID: 29246870 DOI: 10.1016/j.mito.2017.12.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 10/12/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023]
Abstract
Brain injuries in multiple sclerosis (MS) involve immunopathological, structural and metabolic defects on myelin sheath, oligodendrocytes (OLs), axons and neurons suggesting that different cellular mechanisms ultimately result in the formation of MS plaques, demyelination, inflammation and brain damage. Bioenergetics, oxygen and ion metabolism dominate the metabolic and biochemical pathways that maintain neuronal viability and impulse transmission which directly or indirectly point to mitochondrial integrity and adenosine triphosphate (ATP) availability indicating the involvement of mitochondria in the pathogenesis of MS. Loss of myelin proteins including myelin basic protein (MBP), proteolipid protein (PLP), myelin associated glycoprotein (MAG), myelin oligodendrocyte glycoproetin (MOG), 2, 3,-cyclic nucleotide phosphodiestarase (CNPase); microglia and microphage activation, oligodendrocyte apoptosis as well as expression of inducible nitric oxide synthase (i-NOS) and myeloperoxidase activities have been implicated in a subset of Balo's type and relapsing remitting MS (RRMS) lesions indicating the involvement of metabolic defects and oxidative stress in MS. Here, we provide an insighting review of defects in cellular metabolism including energy, oxygen and metal metabolism in MS as well as the relevance of animal models of MS in understanding the molecular, biochemical and cellular mechanisms of MS pathogenesis. Additionally, we also discussed the potential for mitochondrial targets and antioxidant protection for therapeutic benefits in MS.
Collapse
Affiliation(s)
- Reginald C Adiele
- Department of Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada; Cameco MS Neuroscience Research Center, Saskatoon City Hospital, Saskatoon, SK, Canada; Department of Public Health, Concordia University of Edmonton, Edmonton, AB, Canada.
| | - Chiedukam A Adiele
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Nigeria
| |
Collapse
|
157
|
Dendrou CA, Fugger L. Immunomodulation in multiple sclerosis: promises and pitfalls. Curr Opin Immunol 2017; 49:37-43. [PMID: 28926740 DOI: 10.1016/j.coi.2017.08.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 08/26/2017] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) afflicts about 2.5 million people globally and poses a major personal and socioeconomic burden. The recognition of MS as an inflammatory disease, characterized by infiltration of immune cells into the central nervous system, has spurred research into the autoimmune etiology of the condition and has provided the rationale for its treatment through immunomodulation. Experience with immunotherapies in MS to date has suggested a disparity between the observed immune cell infiltration and the progressive loss of neurons. However, recent clinical efforts are providing new insights into progressive MS that once again place the immune system at center stage. This article reviews the main mechanisms of MS immunopathogenesis, and the benefits, risks and challenges of immunomodulatory treatments for the disease.
Collapse
Affiliation(s)
- Calliope A Dendrou
- Nuffield Department of Medicine, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Lars Fugger
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology and MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK.
| |
Collapse
|
158
|
Kim MJ, Lim JY, Park SA, Park SI, Kim WS, Ryu CH, Jeun SS. Effective combination of methylprednisolone and interferon β-secreting mesenchymal stem cells in a model of multiple sclerosis. J Neuroimmunol 2017; 314:81-88. [PMID: 29224961 DOI: 10.1016/j.jneuroim.2017.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 01/07/2023]
Abstract
Methylprednisolone (MP) has been recommended as a standard drug in MS therapies. We previously demonstrated that IFNβ-secreting human bone marrow-derived mesenchymal stem cells (MSCs-IFNβ) exert immunomodulatory effects in experimental autoimmune encephalomyelitic (EAE) mice. In this study, we evaluated whether a combined treatment of MP and MSCs-IFNβ had enhanced therapeutic effects on EAE mice. The combination treatment resulted in enhanced immunomodulatory effects, including reduced production of pro-inflammatory cytokines and increased production of anti-inflammatory cytokines. Thus, our results provide a framework for designing novel experimental protocols to enhance the therapeutic effects of existing MS treatments.
Collapse
Affiliation(s)
- Mi Jin Kim
- Department of Biomedicine & Health Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Yeon Lim
- Postech-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Soon A Park
- Department of Biomedicine & Health Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang In Park
- Institute of Catholic Integrative Medicine (ICIM), Incheon St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Won Shik Kim
- Department of Clinical Pathology Laboratory Science, Daejeon Health Institute of Technology, Daejeon, Republic of Korea
| | - Chung Heon Ryu
- Department of Clinical Pathology Laboratory Science, Daejeon Health Institute of Technology, Daejeon, Republic of Korea.
| | - Sin-Soo Jeun
- Department of Biomedicine & Health Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Neurosurgery, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
159
|
Tapeinou A, Giannopoulou E, Simal C, Hansen BE, Kalofonos H, Apostolopoulos V, Vlamis-Gardikas A, Tselios T. Design, synthesis and evaluation of an anthraquinone derivative conjugated to myelin basic protein immunodominant (MBP 85-99) epitope: Towards selective immunosuppression. Eur J Med Chem 2017; 143:621-631. [PMID: 29216561 DOI: 10.1016/j.ejmech.2017.11.063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/14/2017] [Accepted: 11/22/2017] [Indexed: 02/09/2023]
Abstract
Anthraquinone type compounds, especially di-substituted amino alkylamino anthraquinones have been widely studied as immunosuppressants. The anthraquinone ring is part of mitoxandrone that has been used for the treatment of multiple sclerosis (MS) and several types of tumors. A desired approach for the treatment of MS would be the immunosuppression and elimination of specific T cells that are responsible for the induction of the disease. Herein, the development of a peptide compound bearing an anthraquinone derivative with the potential to specifically destroy the encephalitogenic T cells responsible for the onset of MS is described. The compound consists of the myelin basic protein (MBP) 85-99 immunodominant epitope (MBP85-99) coupled to an anthraquinone type molecule (AQ) via a disulfide (S-S) and 6 amino hexanoic acid (Ahx) residues (AQ-S-S-(Ahx)6MBP85-99). AQ-S-S-(Ahx)6MBP85-99 could bind to HLA II DRB1*-1501 antigen with reasonable affinity (IC50 of 56 nM) The compound was localized to the nucleus of Jurkat cells (an immortalized line of human T lymphocytes) 10 min after its addition to the medium and resulted in lowered Bcl-2 levels (apoptosis). Entrance of the compound was abolished when cells were pre-treated with cisplatin, an inhibitor of thioredoxin reductase. Accordingly, levels of free thiols were elevated in the culture supernatants of Jurkat cells exposed to N-succinimidyl 3-(2-pyridyldithio) propionate coupled to (Ahx)6MBP85-99 via a disulphide (SPDP-S-S-(Ahx)6MBP85-99) but returned to normal after exposure to cisplatin. These results raise the possibility of AQ-S-S-(Ahx)6MBP85-99 being used as an eliminator of encephalitogenic T cells via implication of the thioredoxin system for the generation of the toxic, thiol-containing moiety (AQ-SH). Future experiments would ideally determine whether SPDP-S-S-(Ahx)6MBP85-99 could incorporate into HLA II DRB1*-1501 tetramers and neutralize encephalitogenic T cell lines sensitized to MBP85-99.
Collapse
Affiliation(s)
- Anthi Tapeinou
- Department of Chemistry, University of Patras, GR-26504, Rion, Greece
| | - Efstathia Giannopoulou
- Clinical Oncology Laboratory, University Hospital of Patras, Patras Medical School, GR-26504, Rion, Greece
| | - Carmen Simal
- Department of Chemistry, University of Patras, GR-26504, Rion, Greece
| | - Bjarke E Hansen
- Institute for Inflammation Research, Department of Infectious Diseases and Rheumatology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, DK-2100, Copenhagen, Denmark
| | - Haralabos Kalofonos
- Clinical Oncology Laboratory, University Hospital of Patras, Patras Medical School, GR-26504, Rion, Greece
| | - Vasso Apostolopoulos
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | | | - Theodore Tselios
- Department of Chemistry, University of Patras, GR-26504, Rion, Greece.
| |
Collapse
|
160
|
Ewanchuk BW, Allan ERO, Warren AL, Ramachandran R, Yates RM. The cooling compound icilin attenuates autoimmune neuroinflammation through modulation of the T-cell response. FASEB J 2017; 32:1236-1249. [PMID: 29114087 DOI: 10.1096/fj.201700552r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The synthetic supercooling drug, icilin, and its primary receptor target, the cation channel transient receptor potential (TRP) melastatin-8 (TRPM8), have been described as potent negative regulators of inflammation in the colon. The aim of this study was to determine whether the anti-inflammatory action of icilin could potentially be used to treat autoimmune neuroinflammatory disorders, such as multiple sclerosis (MS). During experimental autoimmune encephalomyelitis (EAE)-a CD4+ T cell-driven murine model of MS-we found that both wild-type (WT) and TRPM8-deficient EAE mice were protected from disease progression during icilin treatment, as evidenced by delays in clinical onset and reductions in neuroinflammation. In vitro, icilin potently inhibited the proliferation of murine and human CD4+ T cells, with the peripheral expansion of autoantigen-restricted T cells similarly diminished by the administration of icilin in mice. Attenuation of both TRPM8-/- and TRP ankyrin-1-/- T-cell proliferation by icilin was consistent with the WT phenotype, which suggests a mechanism that is independent of these channels. In addition, icilin treatment altered the expressional profile of activated CD4+ T cells to one that was indicative of restricted effector function and limited neuroinflammatory potential. These findings identify a potent anti-inflammatory role for icilin in lymphocyte-mediated neuroinflammation and highlight clear pleiotropic effects of the compound beyond classic TRP channel activation.-Ewanchuk, B. W., Allan, E. R. O., Warren, A. L., Ramachandran, R., Yates, R. M. The cooling compound icilin attenuates autoimmune neuroinflammation through modulation of the T-cell response.
Collapse
Affiliation(s)
- Benjamin W Ewanchuk
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Euan R O Allan
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Amy L Warren
- Department of Veterinary Clinical and Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Robin M Yates
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
161
|
Gajofatto A. Spotlight on siponimod and its potential in the treatment of secondary progressive multiple sclerosis: the evidence to date. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:3153-3157. [PMID: 29138536 PMCID: PMC5679692 DOI: 10.2147/dddt.s122249] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Siponimod (BAF312) is a synthetic molecule belonging to the sphingosine-1-phosphate (S1P) modulator family, which has putative neuroprotective properties and well-characterized immunomodulating effects mediated by sequestration of B and T cells in secondary lymphoid organs. Compared to fingolimod (ie, precursor of the S1P modulators commercially available for the treatment of relapsing–remitting [RR] multiple sclerosis [MS]), siponimod exhibits selective affinity for types 1 and 5 S1P receptor, leading to a lower risk of adverse events that are mainly induced by S1P3 receptor activation, such as bradycardia and vasoconstriction. In addition, S1P1 and S1P5 receptors are expressed by neurons and glia and could mediate a possible neuroprotective effect of the drug. A Phase II clinical trial of siponimod for RR MS showed a significant effect of the active drug compared to placebo on reducing gadolinium-enhancing lesions on brain magnetic resonance imaging (MRI) after 3 months of treatment. In a recently completed Phase III trial, treatment with siponimod was associated with a significant reduction in disability progression in secondary progressive (SP) MS patients compared to placebo. In this article, current evidence supporting siponimod efficacy for SP MS is reviewed.
Collapse
Affiliation(s)
- Alberto Gajofatto
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
162
|
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
|
163
|
Pardo G, Jones DE. The sequence of disease-modifying therapies in relapsing multiple sclerosis: safety and immunologic considerations. J Neurol 2017; 264:2351-2374. [PMID: 28879412 PMCID: PMC5688209 DOI: 10.1007/s00415-017-8594-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/11/2017] [Accepted: 08/12/2017] [Indexed: 12/18/2022]
Abstract
The treatment landscape for relapsing forms of multiple sclerosis (RMS) has expanded considerably over the last 10 years with the approval of multiple new disease-modifying therapies (DMTs), and others in late-stage clinical development. All DMTs for RMS are believed to reduce central nervous system immune-mediated inflammatory processes, which translate into demonstrable improvement in clinical and radiologic outcomes. However, some DMTs are associated with long-lasting effects on the immune system and/or serious adverse events, both of which may complicate the use of subsequent therapies. When customizing a treatment program, a benefit–risk assessment must consider multiple factors, including the efficacy of the DMT to reduce disease activity, the short- and long-term safety and immunologic profiles of each DMT, the criteria used to define switching treatment, and the risk tolerance of each patient. A comprehensive benefit–risk assessment can only be achieved by evaluating the immunologic, safety, and efficacy data for DMTs in the controlled clinical trial environment and the postmarketing clinical practice setting. This review is intended to help neurologists make informed decisions when treating RMS by summarizing the known data for each DMT and raising awareness of the multiple considerations involved in treating people with RMS throughout the entire course of their disease.
Collapse
Affiliation(s)
- Gabriel Pardo
- OMRF Multiple Sclerosis Center of Excellence, Oklahoma Medical Research Foundation, 820 NE 15th Street, Oklahoma City, OK, 73104, USA.
| | - David E Jones
- Department of Neurology, University of Virginia School of Medicine, PO Box 800394, Charlottesville, VA, 22908, USA
| |
Collapse
|
164
|
Lorscheider J, Jokubaitis VG, Spelman T, Izquierdo G, Lugaresi A, Havrdova E, Horakova D, Trojano M, Duquette P, Girard M, Prat A, Grand'Maison F, Grammond P, Pucci E, Boz C, Sola P, Ferraro D, Spitaleri D, Lechner-Scott J, Terzi M, Van Pesch V, Iuliano G, Bergamaschi R, Ramo-Tello C, Granella F, Oreja-Guevara C, Butzkueven H, Kalincik T. Anti-inflammatory disease-modifying treatment and short-term disability progression in SPMS. Neurology 2017; 89:1050-1059. [PMID: 28794248 PMCID: PMC5589791 DOI: 10.1212/wnl.0000000000004330] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 06/15/2017] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate the effect of disease-modifying treatment on short-term disability outcomes in secondary progressive multiple sclerosis (SPMS). METHODS Using MSBase, an international cohort study, we previously validated a highly accurate definition of SPMS. Here, we identified patients in MSBase who were either untreated or treated with a disease-modifying drug when meeting this definition. Propensity score matching was used to select subpopulations with comparable baseline characteristics. Disability outcomes were compared in paired, pairwise-censored analyses adjusted for treatment persistence, visit density, and relapse rates. RESULTS Of the 2,381 included patients, 1,378 patients were matchable (treated n = 689, untreated n = 689). Median pairwise-censored follow-up was 2.1 years (quartiles 1.2-3.8 years). No difference in the risk of 6-month sustained disability progression was observed between the groups (hazard ratio [HR] 0.9, 95% confidence interval [CI] 0.7-1.1, p = 0.27). We also did not find differences in any of the secondary endpoints: risk of reaching Expanded Disability Status Scale (EDSS) score ≥7 (HR 0.6, 95% CI 0.4-1.1, p = 0.10), sustained disability reduction (HR 1.0, 95% CI 0.8-1.3, p = 0.79), or change in disability burden (area under the EDSS-time curve, β = -0.05, p = 0.09). Secondary and sensitivity analyses confirmed the results. CONCLUSIONS Our pooled analysis of the currently available disease-modifying agents used after conversion to SPMS suggests that, on average, these therapies have no substantial effect on relapse-unrelated disability outcomes measured by the EDSS up to 4 years. CLASSIFICATION OF EVIDENCE This study provides Class IV evidence that for patients with SPMS, disease-modifying treatment has no beneficial effect on short-term disability progression.
Collapse
|
165
|
Soelberg Sorensen P. Safety concerns and risk management of multiple sclerosis therapies. Acta Neurol Scand 2017; 136:168-186. [PMID: 27891572 DOI: 10.1111/ane.12712] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2016] [Indexed: 01/13/2023]
Abstract
Currently, more than ten drugs have been approved for treatment of relapsing-remitting multiple sclerosis (MS). Newer treatments may be more effective, but have less favorable safety record. Interferon-β preparations and glatiramer acetate treatment require frequent subcutaneous or intramuscular injections and are only moderately effective, but have very rarely life-threatening adverse effects, whereas teriflunomide and dimethyl fumarate are administered orally and have equal or better efficacy, but have more potentially severe adverse effects. The highly effective therapies fingolimod, natalizumab, daclizumab, and alemtuzumab have more serious adverse effects, some of which may be life-threatening. The choice between drugs should be based on a benefit-risk evaluation and tailored to the individual patient's requirements in a dialogue between the patient and treating neurologist. Patients with average disease activity can choose between dimethyl fumarate and teriflunomide or the "old injectable." Patients with very active MS may choose a more effective drug as the initial treatment. In case of side effects on one drug, switch to another drug can be tried. Suboptimal effect of the first drug indicates escalation to a highly efficacious drug. A favorable benefit-risk balance can be maintained by appropriate patient selection and appropriate risk management on therapy. New treatments will within the coming 1-2 years change our current treatment algorithm for relapsing-remitting MS.
Collapse
Affiliation(s)
- P. Soelberg Sorensen
- Department of Neurology; Danish Multiple Sclerosis Center; University of Copenhagen; Rigshospitalet; Copenhagen Denmark
| |
Collapse
|
166
|
Subramaniam V, Chuang G, Xia H, Burn B, Bradley J, Maderdrut JL, Coy DH, Varner KJ. Pituitary adenylate cyclase-activating polypeptide (PACAP) protects against mitoxantrone-induced cardiac injury in mice. Peptides 2017; 95:25-32. [PMID: 28720396 PMCID: PMC5568240 DOI: 10.1016/j.peptides.2017.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/06/2017] [Accepted: 07/11/2017] [Indexed: 12/30/2022]
Abstract
Mitoxantrone (MXT) is an androstenedione that is used to treat cancers and progressive forms of multiple sclerosis; however, its use is limited by its cardiotoxicity. Pituitary adenylate cyclase activating polypeptide (PACAP) is a member of the secretin/growth hormone-releasing hormone/vasoactive intestinal peptide family and has many functions, including cytoprotection and immunosuppression. We tested the hypothesis that PACAP can protect against MXT-induced cardiotoxicity in mice. Female BALB/c mice were treated once weekly for 4 weeks with saline (n=14) or MXT (3mg/kg, i.p.; n=14). Half of the mice in each group received PACAP (10μg, i.p.) 1h before and 24 and 48h after MXT, while the remaining mice received injections of saline on the same schedule. Echocardiography was used to assess cardiac structure and function. In mice treated with MXT and saline, body weight was significantly reduced after the third dose of MXT. PACAP significantly attenuated the reduction in body weight; however, the weights did not return to control level. Compared to controls, MXT-treated mice had significantly increased left ventricular (LV) diameter and LV volume and decreased LV posterior wall thickness. Fractional shortening (FS) and ejection fraction (EF) were also significantly decreased. Treatment with PACAP prevented MXT-induced LV dilation and significantly attenuated the reductions in FS and EF, although FS and EF did not return to control level. PACAP38 did not prevent MXT-induced decreases in LV posterior wall thickness. MXT dose-dependently decreased the viability of cultured U937 (human leukemia) cells; PACAP did not protect cultured U937 cells from MXT-mediated cell death. In conclusion, PACAP can attenuate MXT-mediated LV dilation and dysfunction in mice.
Collapse
Affiliation(s)
- Venkat Subramaniam
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States
| | - Gin Chuang
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States
| | - Huijing Xia
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States; Cardiovascular Center of Excellence, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States
| | - Brendan Burn
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States
| | - Jessica Bradley
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States; Cardiovascular Center of Excellence, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States
| | - Jerome L Maderdrut
- Peptide Research Laboratory, Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112-2699, United States
| | - David H Coy
- Peptide Research Laboratory, Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112-2699, United States
| | - Kurt J Varner
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States; Cardiovascular Center of Excellence, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States.
| |
Collapse
|
167
|
Multiple Sclerosis: Immunopathology and Treatment Update. Brain Sci 2017; 7:brainsci7070078. [PMID: 28686222 PMCID: PMC5532591 DOI: 10.3390/brainsci7070078] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 02/07/2023] Open
Abstract
The treatment of multiple sclerosis (MS) has changed over the last 20 years. All immunotherapeutic drugs target relapsing remitting MS (RRMS) and it still remains a medical challenge in MS to develop a treatment for progressive forms. The most common injectable disease-modifying therapies in RRMS include β-interferons 1a or 1b and glatiramer acetate. However, one of the major challenges of injectable disease-modifying therapies has been poor treatment adherence with approximately 50% of patients discontinuing the therapy within the first year. Herein, we go back to the basics to understand the immunopathophysiology of MS to gain insights in the development of new improved drug treatments. We present current disease-modifying therapies (interferons, glatiramer acetate, dimethyl fumarate, teriflunomide, fingolimod, mitoxantrone), humanized monoclonal antibodies (natalizumab, ofatumumb, ocrelizumab, alentuzumab, daclizumab) and emerging immune modulating approaches (stem cells, DNA vaccines, nanoparticles, altered peptide ligands) for the treatment of MS.
Collapse
|
168
|
Nandoskar A, Raffel J, Scalfari AS, Friede T, Nicholas RS. Pharmacological Approaches to the Management of Secondary Progressive Multiple Sclerosis. Drugs 2017; 77:885-910. [PMID: 28429241 DOI: 10.1007/s40265-017-0726-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
It is well recognised that the majority of the impact of multiple sclerosis (MS), both personal and societal, arises in the progressive phase where disability accumulates inexorably. As such, progressive MS (PMS) has been the target of pharmacological therapies for many years. However, there are no current licensed treatments for PMS. This stands in marked contrast to relapsing remitting MS (RRMS) where trials have resulted in numerous licensed therapies. PMS has proven to be a more difficult challenge compared to RRMS and this review focuses on secondary progressive MS (SPMS), where relapses occur before the onset of gradual, irreversible disability, and not primary progressive MS where disability accumulation occurs without prior relapses. Although there are similarities between the two forms, in both cases pinpointing when PMS starts is difficult in a condition in which disability can vary from day to day. There is also an overlap between the pathology of relapsing and progressive MS and this has contributed to the lack of well-defined outcomes, both surrogates and clinically relevant outcomes in PMS. In this review, we used the search term 'randomised controlled clinical drug trials in secondary progressive MS' in publications since 1988 together with recently completed trials where results were available. We found 34 trials involving 21 different molecules, of which 38% were successful in reaching their primary outcome. In general, the trials were well designed (e.g. double blind) with sample sizes ranging from 35 to 1949 subjects. The majority were parallel group, but there were also multi-arm and multidose trials as well as the more recent use of adaptive designs. The disability outcome most commonly used was the Expanded Disability Status Scale (EDSS) in all phases, but also magnetic resonance imaging (MRI)-measured brain atrophy has been utilised as a surrogate endpoint in phase II studies. The majority of the treatments tested in SPMS over the years were initially successful in RRMS. This has a number of implications in terms of targeting SPMS, but principally implies that the optimal strategy to target SPMS is to utilise the prodrome of relapses to initiate a therapy that will aim to both prevent progression and slow its accumulation. This approach is in agreement with the early targeting of MS but requires treatments that are both effective and safe if it is to be used before disability is a major problem. Recent successes will hopefully result in the first licensed therapy for PMS and enable us to test this approach.
Collapse
Affiliation(s)
- A Nandoskar
- Wolfson Neuroscience Laboratories, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, 160 Du Cane Road, London, W12 0NN, UK
| | - J Raffel
- Wolfson Neuroscience Laboratories, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, 160 Du Cane Road, London, W12 0NN, UK
| | - A S Scalfari
- Wolfson Neuroscience Laboratories, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, 160 Du Cane Road, London, W12 0NN, UK
| | - T Friede
- Department of Medical Statistics, University Medical Center Göttingen, Humboltallee 32, 37073, Göttingen, Germany
| | - R S Nicholas
- Wolfson Neuroscience Laboratories, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, 160 Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
169
|
Yong H, Chartier G, Quandt J. Modulating inflammation and neuroprotection in multiple sclerosis. J Neurosci Res 2017; 96:927-950. [PMID: 28580582 DOI: 10.1002/jnr.24090] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/17/2017] [Accepted: 05/04/2017] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is a neurological disorder of the central nervous system with a presentation and disease course that is largely unpredictable. MS can cause loss of balance, impaired vision or speech, weakness and paralysis, fatigue, depression, and cognitive impairment. Immunomodulation is a major target given the appearance of focal demyelinating lesions in myelin-rich white matter, yet progression and an increasing appreciation for gray matter involvement, even during the earliest phases of the disease, highlights the need to afford neuroprotection and limit neurodegenerative processes that correlate with disability. This review summarizes key aspects of MS pathophysiology and histopathology with a focus on neuroimmune interactions in MS, which may facilitate neurodegeneration through both direct and indirect mechanisms. There is a focus on processes thought to influence disease progression and the role of oxidative stress and mitochondrial dysfunction in MS. The goals and efficacy of current disease-modifying therapies and those in the pipeline are discussed, highlighting recent advances in our understanding of pathways mediating disease progression to identify and translate both immunomodulatory and neuroprotective therapeutics from the bench to the clinic.
Collapse
Affiliation(s)
- Heather Yong
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gabrielle Chartier
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jacqueline Quandt
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
170
|
Abstract
Multiple sclerosis (MS) is the most common disabling neurologic disease of young adults. There are now 16 US Food and Drug Administration (FDA)-approved disease-modifying therapies for MS as well as a cohort of other agents commonly used in practice when conventional therapies prove inadequate. This article discusses approved FDA therapies as well as commonly used practice-based therapies for MS, as well as those therapies that can be used in patients attempting to become pregnant, or in patients with an established pregnancy, who require concomitant treatment secondary to recalcitrant disease activity.
Collapse
|
171
|
Correale J, Gaitán MI, Ysrraelit MC, Fiol MP. Progressive multiple sclerosis: from pathogenic mechanisms to treatment. Brain 2017; 140:527-546. [PMID: 27794524 DOI: 10.1093/brain/aww258] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 08/18/2016] [Indexed: 12/30/2022] Open
Abstract
During the past decades, better understanding of relapsing-remitting multiple sclerosis disease mechanisms have led to the development of several disease-modifying therapies, reducing relapse rates and severity, through immune system modulation or suppression. In contrast, current therapeutic options for progressive multiple sclerosis remain comparatively disappointing and challenging. One possible explanation is a lack of understanding of pathogenic mechanisms driving progressive multiple sclerosis. Furthermore, diagnosis is usually retrospective, based on history of gradual neurological worsening with or without occasional relapses, minor remissions or plateaus. In addition, imaging methods as well as biomarkers are not well established. Magnetic resonance imaging studies in progressive multiple sclerosis show decreased blood-brain barrier permeability, probably reflecting compartmentalization of inflammation behind a relatively intact blood-brain barrier. Interestingly, a spectrum of inflammatory cell types infiltrates the leptomeninges during subpial cortical demyelination. Indeed, recent magnetic resonance imaging studies show leptomeningeal contrast enhancement in subjects with progressive multiple sclerosis, possibly representing an in vivo marker of inflammation associated to subpial demyelination. Treatments for progressive disease depend on underlying mechanisms causing central nervous system damage. Immunity sheltered behind an intact blood-brain barrier, energy failure, and membrane channel dysfunction may be key processes in progressive disease. Interfering with these mechanisms may provide neuroprotection and prevent disability progression, while potentially restoring activity and conduction along damaged axons by repairing myelin. Although most previous clinical trials in progressive multiple sclerosis have yielded disappointing results, important lessons have been learnt, improving the design of novel ones. This review discusses mechanisms involved in progressive multiple sclerosis, correlations between histopathology and magnetic resonance imaging studies, along with possible new therapeutic approaches.
Collapse
|
172
|
Sartori A, Fantini J, Manganotti P. How far away from having an effective treatment option for progressive multiple sclerosis are we? Expert Opin Pharmacother 2017; 18:953-955. [DOI: 10.1080/14656566.2017.1326909] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Arianna Sartori
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Azienda Sanitaria Universitaria Integrata di Trieste, Neurology Unit – Cattinara Hospital, Trieste, Italy
| | - Jacopo Fantini
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Azienda Sanitaria Universitaria Integrata di Trieste, Neurology Unit – Cattinara Hospital, Trieste, Italy
| | - Paolo Manganotti
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Azienda Sanitaria Universitaria Integrata di Trieste, Neurology Unit – Cattinara Hospital, Trieste, Italy
| |
Collapse
|
173
|
Neil S, Huh J, Baronas V, Li X, McFarland HF, Cherukuri M, Mitchell JB, Quandt JA. Oral administration of the nitroxide radical TEMPOL exhibits immunomodulatory and therapeutic properties in multiple sclerosis models. Brain Behav Immun 2017; 62:332-343. [PMID: 28238951 PMCID: PMC5496657 DOI: 10.1016/j.bbi.2017.02.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 02/13/2017] [Accepted: 02/22/2017] [Indexed: 01/01/2023] Open
Abstract
Therapies with both immunomodulatory and neuroprotective properties are thought to have the greatest promise in reducing the severity and progression of multiple sclerosis (MS). Several reactive oxygen (ROS) and reactive nitrogen species (RNS) are implicated in inflammatory-mediated damage to the central nervous system (CNS) in MS and its animal model, experimental autoimmune encephalomyelitis (EAE). TEMPOL (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl) is a stable nitroxide radical with potent antioxidant activity. The goal of our studies was to investigate the immunomodulatory effects and therapeutic potential of orally-delivered TEMPOL in the mouse EAE model. Mice receiving TEMPOL chow ad libitum for 2weeks prior to induction of active EAE showed delayed onset and reduced incidence of disease compared to control-fed animals. Reduced disease severity was associated with limited microglial activation and fewer inflammatory infiltrates. TEMPOL's effects were immunomodulatory, not immunosuppressive: T cells produced less interferon-γ and tumor necrosis factor-α, and TEMPOL-fed mice exhibited a shift towards TH2-type antibody responses. Both myeloid and myeloid-dendritic cells of TEMPOL-fed EAE animals had significantly lower levels of MHC class II expression than controls; CD40 was also significantly reduced. TEMPOL administration was associated with an enrichment of CD8+ T cell populations and CD4+FoxP3+ regulatory populations. TEMPOL reduced the severity of clinical disease when administered after the induction of disease, and also after the onset of clinical symptoms. To exclude effects on T cell priming in vivo, TEMPOL was tested with the passive transfer of encephalitogenic T cells and was found to reduce the incidence and peak severity of disease. Protection was associated with reduced infiltrates and a relative sparing of neurofilaments and axons. The ability of oral TEMPOL to reduce inflammation and axonal damage and loss demonstrate both anti-inflammatory and protective properties, with significant promise for the treatment of MS and related neurological disorders.
Collapse
Affiliation(s)
- Sarah Neil
- University of British Columbia, Department of Pathology & Laboratory Medicine, Vancouver, Canada
| | - Jaebong Huh
- Neuroimmunology Branch, NINDS, NIH, Bethesda, MD 20892 USA
| | - Victoria Baronas
- University of British Columbia, Department of Pathology & Laboratory Medicine, Vancouver, Canada
| | - Xinhui Li
- Neuroimmunology Branch, NINDS, NIH, Bethesda, MD 20892 USA
| | | | | | | | - Jacqueline A. Quandt
- University of British Columbia, Department of Pathology & Laboratory Medicine, Vancouver, Canada,To whom correspondence should be addressed: University of British Columbia, Department of Pathology & Laboratory Medicine, G227-2211 Wesbrook Mall, Vancouver, B.C. V6T 2B5, Canada,
| |
Collapse
|
174
|
McNamara C, Sugrue G, Murray B, MacMahon PJ. Current and Emerging Therapies in Multiple Sclerosis: Implications for the Radiologist, Part 1-Mechanisms, Efficacy, and Safety. AJNR Am J Neuroradiol 2017; 38:1664-1671. [PMID: 28408630 DOI: 10.3174/ajnr.a5147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Imaging for the diagnosis and follow-up of patients with suspected or confirmed multiple sclerosis is a common scenario for many general radiologists and subspecialty neuroradiologists. The field of MS therapeutics has rapidly evolved with multiple new agents now being used in routine clinical practice. To provide an informed opinion in discussions concerning newer MS agents, radiologists must have a working understanding of the strengths and limitations of the various novel therapies. The role of imaging in MS has advanced beyond monitoring and surveillance of disease activity to include treatment complications. An understanding of the new generation of MS drugs in conjunction with the key role that MR imaging plays in the detection of disease progression, opportunistic infections, and drug-related adverse events is of vital importance to the radiologist and clinical physician alike. Radiologists are in a unique position to detect many of the described complications well in advance of clinical symptoms. Part 1 of this review outlines recent developments in the treatment of MS and discusses the published clinical data on the efficacy and safety of the currently approved and emerging therapies in this condition as they apply to the radiologist. Part 2 will cover pharmacovigilance and the role the neuroradiologist plays in monitoring patients for signs of opportunistic infection and/or disease progression.
Collapse
Affiliation(s)
- C McNamara
- From the Departments of Radiology (C.M., G.S., P.J.M.)
| | - G Sugrue
- From the Departments of Radiology (C.M., G.S., P.J.M.)
| | - B Murray
- Neurology (B.M.), Mater Misericordiae University Hospital, Dublin, Ireland
| | - P J MacMahon
- From the Departments of Radiology (C.M., G.S., P.J.M.)
| |
Collapse
|
175
|
Comi G, Radaelli M, Soelberg Sørensen P. Evolving concepts in the treatment of relapsing multiple sclerosis. Lancet 2017; 389:1347-1356. [PMID: 27889192 DOI: 10.1016/s0140-6736(16)32388-1] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 10/18/2016] [Accepted: 10/20/2016] [Indexed: 11/16/2022]
Abstract
In the past 20 years the treatment scenario of multiple sclerosis has radically changed. The increasing availability of effective disease-modifying therapies has shifted the aim of therapeutic interventions from a reduction in relapses and disability accrual, to the absence of any sign of clinical or MRI activity. The choice for therapy is increasingly complex and should be driven by an appropriate knowledge of the mechanisms of action of the different drugs and of their risk-benefit profile. Because the relapsing phase of the disease is characterised by inflammation, treatment should be started as early as possible and aim to re-establish the normal complex interactions in the immune system. Before starting a treatment, neurologists should carefully consider the state of the disease, its prognostic factors and comorbidities, the patient's response to previous treatments, and whether the patient is likely to accept treatment-related risks in order to maximise benefits and minimise risks. Early detection of suboptimum responders, thanks to accurate clinical monitoring, will allow clinicians to redesign treatment strategies where necessary.
Collapse
Affiliation(s)
- Giancarlo Comi
- Department of Neurology and Institute of Experimental Neurology, San Raffaele Hospital, Milan, Italy.
| | - Marta Radaelli
- Department of Neurology and Institute of Experimental Neurology, San Raffaele Hospital, Milan, Italy
| | - Per Soelberg Sørensen
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
176
|
Abstract
Most of the current therapies, as well as many of the clinical trials, for multiple sclerosis (MS) target the inflammatory autoimmune processes, but less than 20% of all clinical trials investigate potential therapies for the chronic progressive disease stage of MS. The latter is responsible for the steadily increasing disability in many patients, and there is an urgent need for novel therapies that protect nervous system tissue and enhance axonal growth and/or remyelination. As outlined in this review, solid pre-clinical data suggest neutralization of the neurite outgrowth inhibitor Nogo-A as a potential new way to achieve both axonal and myelin repair. Several phase I clinical studies with anti-Nogo-A antibodies have been conducted in different disease paradigms including MS and spinal cord injury. Data from spinal cord injury and amyotrophic lateral sclerosis (ALS) trials accredit a good safety profile of high doses of anti-Nogo-A antibodies administered intravenously or intrathecally. An antibody against a Nogo receptor subunit, leucine rich repeat and immunoglobulin-like domain-containing protein 1 (LINGO-1), was recently shown to improve outcome in patients with acute optic neuritis in a phase II study. Nogo-A-suppressing antibodies could be novel drug candidates for the relapsing as well as the progressive MS disease stage. In this review, we summarize the available pre-clinical and clinical evidence on Nogo-A and elucidate the potential of Nogo-A-antibodies as a therapy for progressive MS.
Collapse
|
177
|
Consensus statement on the treatment of multiple sclerosis by the Spanish Society of Neurology in 2016. NEUROLOGÍA (ENGLISH EDITION) 2017. [DOI: 10.1016/j.nrleng.2016.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
178
|
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
|
179
|
Feng JJ, Ontaneda D. Treating primary-progressive multiple sclerosis: potential of ocrelizumab and review of B-cell therapies. Degener Neurol Neuromuscul Dis 2017; 7:31-45. [PMID: 30050376 PMCID: PMC6053100 DOI: 10.2147/dnnd.s100096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multiple sclerosis (MS) therapy has evolved rapidly with an increased availability of several immunomodulating therapies over the past two decades. Disease-modifying therapies have proven to be effective in treating relapse-remitting MS (RRMS). However, clinical trials involving some of the same agents for secondary-progressive and primary-progressive MS (SPMS and PPMS) have been largely negative. The pathogenesis of progressive MS remains unclear, but B-cells may play a significant role in chronic compartmentalized inflammation, likely contributing to disease progression. Biologics targeted at B-cells, such as rituximab, are effective in treating RRMS. Ocrelizumab is a humanized monoclonal antibody to CD20+ B-cells that has shown positive results in PPMS with a significant reduction in disease progression. This review aims to discuss in detail the involvement of B-cells in MS pathogenesis, current progress of currently available and investigational biologics, with focus on ocrelizumab, and future prospects for B-cell therapy in PPMS.
Collapse
Affiliation(s)
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Cleveland Clinic, Cleveland, OH, USA,
| |
Collapse
|
180
|
Brochet B, Deloire MSA, Perez P, Loock T, Baschet L, Debouverie M, Pittion S, Ouallet JC, Clavelou P, de Sèze J, Collongues N, Vermersch P, Zéphir H, Castelnovo G, Labauge P, Lebrun C, Cohen M, Ruet A, PROMESS study investigators. Double-Blind Controlled Randomized Trial of Cyclophosphamide versus Methylprednisolone in Secondary Progressive Multiple Sclerosis. PLoS One 2017; 12:e0168834. [PMID: 28045953 PMCID: PMC5207788 DOI: 10.1371/journal.pone.0168834] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 12/04/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Therapeutic options are limited in secondary progressive multiple sclerosis (SPMS). Open-label studies suggested efficacy of monthly IV cyclophosphamide (CPM) without induction for delaying progression but no randomized trial was conducted so far. OBJECTIVE To compare CPM to methylprednisolone (MP) in SPMS. METHODS Randomized, double-blind clinical trial on two parallel groups. Patient with SPMS, with a documented worsening of the Expanded Disability Status Scale (EDSS) score during the last year and an EDSS score between 4·0 and 6·5 were recruited and received one intravenous infusion of treatment (CPM: 750 mg /m2 body surface area-MP: 1g) every four weeks for one year, and every eight weeks for the second year. The primary endpoint was the time to EDSS deterioration, when confirmed sixteen weeks later, analyzed using a Cox model. RESULTS Due to recruitment difficulties, the study was terminated prematurely after 138 patients were included (CPM, n = 72; MP, n = 66). In the CPM group, 33 patients stopped treatment prematurely, mainly due to tolerability, compared with 22 in the MP group. Primary endpoint: the hazard ratio for EDSS deterioration in the CPM in comparison with the MP group was 0.61 [95% CI: 0·31-1·22](p = 0·16). According to the secondary multistate model analysis, patients in the CPM group were 2.2 times more likely ([1·14-4.29]; p = 0.02) to discontinue treatment than those in the MP group and 2.7 times less likely (HR = 0.37, 95% CI: 0.17-0.84; p = 0.02) to experience disability progression when they did not stop treatment prematurely. Safety profile was as expected. CONCLUSION Although the primary end-point was negative, secondary analysis suggested that CPM decreases the risk of progression in SPMS, but its use may be limited by low tolerability. TRIAL REGISTRATION Clinicaltrials.gov NCT00241254.
Collapse
Affiliation(s)
- Bruno Brochet
- Service de Neurologie et INSERM-CHU CIC-P 0005, CHU de Bordeaux, Bordeaux, France
- INSERM U 1215, Université de Bordeaux, Bordeaux, France
- * E-mail:
| | | | - Paul Perez
- Unité de Soutien Méthodologique à la Recherche Clinique et Epidémiologique, Pôle de Santé Publique, CHU de Bordeaux, Bordeaux France
| | - Timothé Loock
- Service de Neurologie et INSERM-CHU CIC-P 0005, CHU de Bordeaux, Bordeaux, France
| | - Louise Baschet
- Unité de Soutien Méthodologique à la Recherche Clinique et Epidémiologique, Pôle de Santé Publique, CHU de Bordeaux, Bordeaux France
| | | | | | | | - Pierre Clavelou
- Service de Neurologie, CHU de Clermont-Ferrand, Clermont-Ferrand, France
| | - Jérôme de Sèze
- Service de Neurologie et CIC INSERM 1434, CHU de Strasbourg, Strasbourg, France
| | - Nicolas Collongues
- Service de Neurologie et CIC INSERM 1434, CHU de Strasbourg, Strasbourg, France
| | - Patrick Vermersch
- Univ. Lille, CHU Lille, LIRIC-INSERM U995, FHU Imminent, Lille, France
| | - Hélène Zéphir
- Univ. Lille, CHU Lille, LIRIC-INSERM U995, FHU Imminent, Lille, France
| | | | - Pierre Labauge
- Service de Neurologie, CHU de Montpellier, Montpellier, France
| | | | - Mikael Cohen
- Service de Neurologie, CHU de Nice, Nice, France
| | - Aurélie Ruet
- Service de Neurologie et INSERM-CHU CIC-P 0005, CHU de Bordeaux, Bordeaux, France
- INSERM U 1215, Université de Bordeaux, Bordeaux, France
| | | |
Collapse
|
181
|
Abstract
Multiple sclerosis (MS) is a chronic disease of the central nervous system (CNS) characterized by loss of motor and sensory function that results from immune-mediated inflammation, demyelination, and subsequent axonal damage. Clinically, most MS patients experience recurrent episodes (relapses) of neurological impairment, but in most cases (60–80%) the course of the disease eventually becomes chronic and progressive, leading to cumulative motor, sensory, and visual disability, and cognitive deficits. The course of the disease is largely unpredictable and its clinical presentation is variable, but its predilection for certain parts of the CNS, which includes the optic nerves, the brain stem, cerebellum, and cervical spinal cord, provides a characteristic constellation of signs and symptoms. Several variants of MS have been nowadays defined with variable immunopathogenesis, course and prognosis. Many new treatments targeting the immune system have shown efficacy in preventing the relapses of MS and have been introduced to its management during the last decade.
Collapse
|
182
|
Comini-Frota ER, Vasconcelos CCF, Mendes MF. Guideline for multiple sclerosis treatment in Brazil: Consensus from the Neuroimmunology Scientific Department of the Brazilian Academy of Neurology. ARQUIVOS DE NEURO-PSIQUIATRIA 2017; 75:57-65. [DOI: 10.1590/0004-282x20160185] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 09/22/2016] [Indexed: 12/17/2022]
Abstract
ABSTRACT Multiple sclerosis has become an ever-increasing challenge to neurologists. With the release of the latest medications on the market, Brazilian neurologists feel divided between following their patients’ evolution in accordance with the strict rules established by the Brazilian Ministry of Health regarding drug distribution, or following disease progression and worsening in accordance with the evidence in the literature. Therefore, a systematic review of the main published treatment guidelines was conducted and an escalating therapy proposed for guiding multiple sclerosis patient treatment in Brazil.
Collapse
|
183
|
Lasek-Bal A, Bartoszek K, Steposz A, Puz P, Bal W, Kazibutowska Z. Efficacy and safety of mitoxantrone use in primary and secondary progressive multiple sclerosis – study site experience based on the therapy of 104 patients. Int J Neurosci 2016; 127:859-863. [DOI: 10.1080/00207454.2016.1269327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Anetta Lasek-Bal
- Department of Neurolog, School of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
- Department of Neurology, Professor Leszek Giec Upper Silesian Medical Centre, Medical University of Silesia Hospital No. 7, Katowice, Poland
| | - Karina Bartoszek
- Department of Neurology, Professor Leszek Giec Upper Silesian Medical Centre, Medical University of Silesia Hospital No. 7, Katowice, Poland
| | - Arkadiusz Steposz
- Department of Neurolog, School of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
- Department of Neurology, Professor Leszek Giec Upper Silesian Medical Centre, Medical University of Silesia Hospital No. 7, Katowice, Poland
| | - Przemyslaw Puz
- Department of Neurolog, School of Health Sciences in Katowice, Medical University of Silesia, Katowice, Poland
- Department of Neurology, Professor Leszek Giec Upper Silesian Medical Centre, Medical University of Silesia Hospital No. 7, Katowice, Poland
| | - Wieslaw Bal
- Department of Radiation Oncology and Chemotherapy, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Zofia Kazibutowska
- Department of Neurology, Professor Leszek Giec Upper Silesian Medical Centre, Medical University of Silesia Hospital No. 7, Katowice, Poland
| |
Collapse
|
184
|
Díaz Sánchez M, Jiménez Hernández M. Tratamiento de las enfermedades desmielinizantes. Esclerosis múltiple. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.med.2016.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
185
|
Gajofatto A, Turatti M, Benedetti MD. Primary progressive multiple sclerosis: current therapeutic strategies and future perspectives. Expert Rev Neurother 2016; 17:393-406. [PMID: 27813441 DOI: 10.1080/14737175.2017.1257385] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic inflammatory condition of the central nervous system with heterogeneous features. Primary progressive (PP) MS is a rare disease subtype characterized by continuous disability worsening from onset. No disease-modifying therapy is currently approved for PP MS due to the negative or inconsistent results of clinical trials conducted on a wide range of interventions, which are reviewed in the present paper. Areas covered: The features and results of randomized trials of disease-modifying treatments for PP MS are discussed, including immunosuppressants, immunomodulators, monoclonal antibodies, and putative neuroprotective agents. Expert commentary: The recent encouraging results of the ocrelizumab trial in PP MS, the first to reach the primary disability endpoint, indicate B cells as a promising therapeutic target to prevent disease progression. Other emerging treatment strategies include cell metabolism modulation and inflammatory pathways inhibition, which are being investigated in several ongoing phase II and III placebo-controlled trials. Future PP MS trials will need to systematically include efficacy endpoints other than physical disability alone, such as cognition, quality of life, advanced MRI measures and molecular biomarkers.
Collapse
Affiliation(s)
- Alberto Gajofatto
- a Department of Neuroscience, Biomedicine and Movement Sciences , University of Verona , Verona , Italy
| | - Marco Turatti
- b Department of Neuroscience , Azienda Ospedaliera Universitaria Integrata Verona , Verona , Italy
| | - Maria Donata Benedetti
- b Department of Neuroscience , Azienda Ospedaliera Universitaria Integrata Verona , Verona , Italy
| |
Collapse
|
186
|
Tourbah A, Lebrun-Frenay C, Edan G, Clanet M, Papeix C, Vukusic S, De Sèze J, Debouverie M, Gout O, Clavelou P, Defer G, Laplaud DA, Moreau T, Labauge P, Brochet B, Sedel F, Pelletier J. MD1003 (high-dose biotin) for the treatment of progressive multiple sclerosis: A randomised, double-blind, placebo-controlled study. Mult Scler 2016; 22:1719-1731. [PMID: 27589059 PMCID: PMC5098693 DOI: 10.1177/1352458516667568] [Citation(s) in RCA: 214] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 07/22/2016] [Accepted: 08/04/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Treatment with MD1003 (high-dose biotin) showed promising results in progressive multiple sclerosis (MS) in a pilot open-label study. OBJECTIVE To confirm the efficacy and safety of MD1003 in progressive MS in a double-blind, placebo-controlled study. METHODS Patients (n = 154) with a baseline Expanded Disability Status Scale (EDSS) score of 4.5-7 and evidence of disease worsening within the previous 2 years were randomised to 12-month MD1003 (100 mg biotin) or placebo thrice daily, followed by 12-month MD1003 for all patients. The primary endpoint was the proportion of patients with disability reversal at month 9, confirmed at month 12, defined as an EDSS decrease of ⩾1 point (⩾0.5 for EDSS 6-7) or a ⩾20% decrease in timed 25-foot walk time compared with the best baseline among screening or randomisation visits. RESULTS A total of 13 (12.6%) MD1003-treated patients achieved the primary endpoint versus none of the placebo-treated patients (p = 0.005). MD1003 treatment also reduced EDSS progression and improved clinical impression of change compared with placebo. Efficacy was maintained over follow-up, and the safety profile of MD1003 was similar to that of placebo. CONCLUSION MD1003 achieves sustained reversal of MS-related disability in a subset of patients with progressive MS and is well tolerated.
Collapse
Affiliation(s)
- Ayman Tourbah
- Department of Neurology and Faculté de Médecine de Reims, CHU de Reims, URCA, Reims, France
| | | | - Gilles Edan
- Service de Neurologie, CHU de Rennes, CICP 1414 INSERM, Rennes, France
| | - Michel Clanet
- Department of Neurology, CHU de Toulouse, Toulouse, France
| | - Caroline Papeix
- Department of Neurology, GH Pitié Salpêtrière, Paris, France
| | - Sandra Vukusic
- Department of Neurology A, Hospices Civils de Lyon, Lyon, France
| | - Jerome De Sèze
- Department of Neurology and Clinical Investigation Center, CHU de Strasbourg, INSERM 1434, Strasbourg, France
| | | | - Olivier Gout
- Department of Neurology, Fondation Ophtalmologique Adolphe de Rothschild, Paris, France
| | - Pierre Clavelou
- Department of Neurology, CHU de Clermont-Ferrand, Clermont-Ferrand, France
| | - Gilles Defer
- Department of Neurology, CHU de Caen, Caen, France
| | - David-Axel Laplaud
- Department of Neurology, CHU de Nantes, CIC015 INSERM, INSERM CR1064, Nantes, France
| | - Thibault Moreau
- Department of Neurology, University Hospital of Dijon, Dijon, France
| | - Pierre Labauge
- Department of Neurology, CHU de Montpellier, Montpellier, France
| | - Bruno Brochet
- Department of Neurology, CHU de Bordeaux, Bordeaux, France
| | | | - Jean Pelletier
- APHM, Hôpital de la Timone, CNRS, CRMBM UMR 7339, CNRS, Department of Neurology and Aix-Marseille Université, Marseille, France
| |
Collapse
|
187
|
Kinner M, Hoepner R, Klotz P, Prehn C, Faissner S, Salmen A, Linker RA, Gold R, Chan A. Immunotherapy Improves Cognitive Function in Secondary Progressive Multiple Sclerosis. CNS Neurosci Ther 2016; 22:1019-1022. [PMID: 27739247 DOI: 10.1111/cns.12652] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/21/2016] [Accepted: 09/22/2016] [Indexed: 12/01/2022] Open
Affiliation(s)
- Markus Kinner
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Robert Hoepner
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Peter Klotz
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Christian Prehn
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Simon Faissner
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Anke Salmen
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany.,Department of Neurology, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ralf A Linker
- Department of Neurology, University of Erlangen, Erlangen, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Andrew Chan
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany.,Department of Neurology, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
188
|
|
189
|
Kretzschmar B, Pellkofer H, Weber MS. The Use of Oral Disease-Modifying Therapies in Multiple Sclerosis. Curr Neurol Neurosci Rep 2016; 16:38. [PMID: 26944956 DOI: 10.1007/s11910-016-0639-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Three oral disease-modifying drugs-fingolimod, teriflunomide, and dimethyl fumarate (DMF)-are available for treatment of relapsing forms of multiple sclerosis (MS). All three agents were approved in the last decade, primarily on the basis of a moderate to substantial reduction in the occurrence of MS relapses and central nervous system lesion formation detected by MRI. In the trials leading to approval, the first oral disease-modifying drug, fingolimod, reduced the annualized relapse rate (ARR) from 0.40 in placebo-treated patients to 0.18 (FREEDOMS) and from 0.33 in patients treated with interferon β1a intramuscularly to 0.16 (TRANSFORMS). Teriflunomide, approved on the basis of the two placebo-controlled trials TEMSO and TOWER, demonstrated a reduction in the ARR from 0.54 to 0.37 and from 0.50 to 0.32 respectively. The latest oral MS medication, approved in 2014, is DMF, which had been used in a different formulation for treatment of psoriasis for decades. In the 2-year DEFINE study, the proportion of patients with a relapse was reduced to 27 %, compared with 46 % in placebo arm, whereas in the CONFIRM trial, the ARR was reduced from 0.40 (placebo) to 0.22 in the DMF-treated group of patients. In this review, we will elucidate the mechanisms of action of these three medications and compare their efficacy, safety, and tolerability as a practical guideline for their use. We will further discuss effects other than relapse reduction these small molecules may exert, including potential activities within the central nervous system, and briefly summarize emerging data on new oral MS drugs in clinical development.
Collapse
Affiliation(s)
- Benedikt Kretzschmar
- Department of Neurology, University Medical Center, 37075, Göttingen, Germany
- Doctor's Office Knaak/Christmann/Wüstenhagen of Neurology and Psychiatry, 34346, Hann. Münden, Germany
| | - Hannah Pellkofer
- Department of Neurology, University Medical Center, 37075, Göttingen, Germany
- Institute of Neuropathology, University Medical Center, 37075, Göttingen, Germany
| | - Martin S Weber
- Department of Neurology, University Medical Center, 37075, Göttingen, Germany.
- Institute of Neuropathology, University Medical Center, 37075, Göttingen, Germany.
- Department of Neuropathology, Department of Neurology, University Medical Center, Georg August University, Robert-Koch-Str. 40, 37099, Göttingen, Germany.
| |
Collapse
|
190
|
[Cell depletion and myoablation for neuroimmunological diseases]. DER NERVENARZT 2016; 87:814-20. [PMID: 27389598 DOI: 10.1007/s00115-016-0156-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND The treatment of autoimmune disorders of the nervous system is based on interventions for the underlying immune phenomena. OBJECTIVE To summarize concepts of cell depletion and myeloablation studied in the context of neuroimmunological disorders. METHOD Evaluation of the available literature on multiple sclerosis as the most widely studied neuroimmunological entity. RESULTS Three concepts have been introduced: classical immunosuppressants, such as azathioprine, mitoxantrone and cyclophosphamide exert general lymphopenic effects and thereby moderately decrease disease activity. Myeloablative regimens combined with autologous hematopoietic stem cell transplantation have a profound and in most cases long-lasting impact on autoimmunity at the cost of potentially life-threatening side effects. Alemtuzumab (anti-CD52), rituximab and ocrelizumab (both anti-CD20) are depleting antibodies directed against certain lymphocyte subsets and substantially ameliorate disease activity in relapsing-remitting multiple sclerosis. Ocrelizumab also shows efficacy in the primary progressive form of multiple sclerosis. CONCLUSIONS Most of the presented cell-depleting and myeloablative therapies are highly effective treatment options but are also accompanied by significant risks. In the context of the increasing number of alternative immunomodulatory options the indications for use should be cautiously considered.
Collapse
|
191
|
Daumer M, Griffith LM, Meister W, Nash RA, Wolinsky JS. Survival, and time to an advanced disease state or progression, of untreated patients with moderately severe multiple sclerosis in a multicenter observational database: relevance for design of a clinical trial for high dose immunosuppressive therapy with autologous hematopoietic stem cell transplantation. Mult Scler 2016; 12:174-9. [PMID: 16629420 DOI: 10.1191/135248506ms1256oa] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Despite prolonged survival, patients with multiple sclerosis (MS) experience considerable morbidity, which adversely impacts quality of life. To assess the risk-benefit of a clinical trial of high dose immunosuppressive therapy with autologous hematopoietic stem cell transplantation for MS, we sought to determine the natural history of the disease in a comparison group of untreated patients. We identified 285 individuals with 2132 combined observation years (median: 5.6 years; 5th to 95th percentile: 1-21 years), with Expanded Disability Status Scale (EDSS) scores of 3.0-5.5 at baseline observation. Disease-related mortality was zero at five years, 5.4% at 10 years, and 22% at 15 years (40 patients contributing to the data point; 95% confidence interval: 4-32%). Risk for progression to advanced disability, defined as an EDSS score of 8, was very low for the subgroup with a baseline EDSS score of 3-3.5; however, for those with a baseline EDSS score of 4-5.5, 3% had advanced disability after two years, 5% after three years, 6% after four years, 12% after five years, and 40% after 10 years. The estimated probability of disease progression, defined as an increase in EDSS score by ≥ 1.0 sustained for at least 180 days, was 5% after one year, 14% after two years, 22% after three years, 38% after five years, 57% after 10 years, and-80% after 20 years of observation. The relevance of these features to the design of the clinical trial is discussed.
Collapse
Affiliation(s)
- M Daumer
- Sylvia Lawry Centre for Multiple Sclerosis Research, Munich, Germany.
| | | | | | | | | |
Collapse
|
192
|
Debouverie M, Vandenberghe N, Morrissey SP, Anxionnat R, Pittion-Vouyovitch S, Vespignani H, Edan G. Predictive parameters of mitoxantrone effectiveness in the treatment of multiple sclerosis. Mult Scler 2016; 10:407-12. [PMID: 15327038 DOI: 10.1191/1352458504ms1066oa] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Introduction: In a number of controlled trials it was established that mitoxantrone has a beneficial effect on disease progression in multiple sclerosis (MS) patients with a worsening disease course. The aim of this study was to investigate the use of mitoxantrone in clinical practice, and especially to describe predictive parameters of its effectiveness under these conditions. Objectives and methods: In a retrospective, open-label mitoxantrone study we analysed 94 MS patients (49) relapsing-remitting MS (RRMS), 41) secondary progressive MS and 10) primary progressive MS) who received monthly 20 mg i.v. mitoxantrone and 1 g i.v. methylprednisolone for six months, and selected as a criterion of effectiveness the percentage of patients with an Expanded Disability Status Scale (EDSS) improvement of at least one point (confirmed after one year) after stopping the treatment. A multivariate analysis was undertaken to assess the predictive value of five parameters on mitoxantrone effectiveness: (1) total number of relapses since disease onset and before treatment, (2) number of relapses within the past 24 months before treatment, (3) number of relapses in separate areas within the past 24 months before treatment, (4) active MRI scans (including Gd-enhanced lesions), and (5) clinical course of MS. Results: During the observation period from 1 January 1997 to 30 May 2000 more than 44) of the patients improved by one point or more on the EDSS (confirmed after one year), 39) remained stable and 17) deteriorated. In patients with a RRMS course three or more relapses within the past 24 months preceding treatment, and at least one Gd-enhancing lesion resulted in a strong relative benefit (i.e., relative risk) of mitoxantrone effectiveness. In contrast, total number of relapses since disease onset had no impact on disease evolution and disability progression. Multivariate analysis revealed the number of relapses in separate areas within the past 24 months before treatment as the strongest predictive parameter (PB/0.001). Conclusion: Mitoxantrone is effective in improving and stabilizing patients with a worsening MS course in routine clinical practice. Several strong predictive parameters of mitoxantrone effectiveness were investigated among which the number of relapses in separate areas within the past 24 months before treatment was found to be the strongest parameter to predict clinical improvement.
Collapse
Affiliation(s)
- M Debouverie
- Department of Neurology, Hôpital Central, Nancy, France.
| | | | | | | | | | | | | |
Collapse
|
193
|
Haas J, Maas-Enriquez M, Hartung HP. Intravenous immunoglobulins in the treatment of relapsing remitting multiple sclerosis - results of a retrospective multicenter observational study over five years. Mult Scler 2016; 11:562-7. [PMID: 16193894 DOI: 10.1191/1352458505ms1224oa] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Use of intravenous immunoglobulins (IVIG) has been recommended for treatment of RRMS if first line therapy with beta-interferon or glatiramer acetate is not tolerated, or if contraindications exist. This consensus recommendation is based on the demonstration of efficacy and tolerability of IVIG in four randomized controlled trials (RCTs). The impact of non-randomized observational trials on evidence-based recommendations for treatment is still under discussion. In order to evaluate the transferability of study results derived from RCTs into a routine practice setting, we carried out a retrospective data analysis on patients with RRMS who had been treated with IVIG during the last five years. Data sets from 308 out of 1122 screened patients were available for analysis. Treatment with IVIG resulted in a 69% reduction of the mean annual relapse rate (ARR) (calculated over two years) from 1.749±1.15 before IVIG treatment to 0.539±0.61 after start of IVIG treatment. Mean expanded disability status scale (EDSS) values remained stable throughout the observation period. The results of this observational study were similar to the results of previous RCTs with IVIG.
Collapse
Affiliation(s)
- J Haas
- Department of Neurology, Jewish Hospital, Berlin, Germany
| | | | | |
Collapse
|
194
|
McFarland HF, Reingold SC. The future of multiple sclerosis therapies: redesigning multiple sclerosis clinical trials in a new therapeutic eraa. Mult Scler 2016; 11:669-76. [PMID: 16320726 DOI: 10.1191/1352458505ms1231oa] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Due to past success in testing and gaining regulatory approval for a variety of therapies in multiple sclerosis (MS), the conduct of future clinical trials has become increasingly problematic. An international workshop has met to discuss the issues facing the MS clinical trial community and to examine possible new strategies for the design of trials. Particular focus has been placed on trials that either avoid the use of a placebo because of ethical considerations or on designs that allow new therapies to be studied more rapidly or with fewer patients than needed in a conventional placebo-controlled trial. The discussions resulting from the workshop should provide a basis for the examination and implementation of innovative clinical trial designs in MS.
Collapse
Affiliation(s)
- H F McFarland
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | | |
Collapse
|
195
|
Luessi F, Zipp F, Witsch E. Dendritic cells as therapeutic targets in neuroinflammation. Cell Mol Life Sci 2016; 73:2425-50. [PMID: 26970979 PMCID: PMC11108452 DOI: 10.1007/s00018-016-2170-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 02/02/2016] [Accepted: 02/25/2016] [Indexed: 12/23/2022]
Abstract
Multiple sclerosis (MS) is the most common chronic inflammatory demyelinating disorder of the central nervous system characterized by infiltration of immune cells and progressive damage to myelin sheaths and neurons. There is still no cure for the disease, but drug regimens can reduce the frequency of relapses and slightly delay progression. Myeloid cells or antigen-presenting cells (APCs) such as dendritic cells (DC), macrophages, and resident microglia, are key players in both mediating immune responses and inducing immune tolerance. Mounting evidence indicates a contribution of these myeloid cells to the pathogenesis of multiple sclerosis and to the effects of treatment, the understanding of which might provide strategies for more potent novel therapeutic interventions. Here, we review recent insights into the role of APCs, with specific focus on DCs in the modulation of neuroinflammation in MS.
Collapse
Affiliation(s)
- Felix Luessi
- Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University of Mainz,Rhine Main Neuroscience Network (rmn2), Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University of Mainz,Rhine Main Neuroscience Network (rmn2), Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Esther Witsch
- Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University of Mainz,Rhine Main Neuroscience Network (rmn2), Langenbeckstrasse 1, 55131, Mainz, Germany.
| |
Collapse
|
196
|
Abstract
Multiple sclerosis (MS) is a complex disease that causes a great deal of disability, especially in the young adult population. There have been several immunomodulatory agents that have been approved by the Food and Drug Administration for MS, including glatiramer acetate, interferon-β 1a and -β 1b, mitoxantrone, and corticosteroids. The effectiveness of these therapies has not been optimal, and drugs, such as monoclonal antibodies, that more selectively target the pathogenetic process of MS have been sought. These agents have their own intrinsic limitations such as systemic inflammatory reactions, induction of neutralizing antiantibodies, and even life-threatening infectious processes. The agent that has been in the forefront of the discussion is natalizumab, a monoclonal antibody (mAb) against α 4 integrin, which shows much promise in suppressing MS activity. However, 3 individuals treated with natalizumab developed a life-threatening infection, progressive multifocal leukoencephalopathy. This article reviews the role of mAbs in the treatment of MS, particularly their advantages over other drugs and their limitations, which have to be overcome for these agents to be at the forefront in the treatment of MS.
Collapse
Affiliation(s)
| | - Jane W. Chan
- University of Kentucky College of Medicine, Lexington,
| |
Collapse
|
197
|
|
198
|
Barry A, Cronin O, Ryan AM, Sweeney B, Yap SM, O'Toole O, Allen AP, Clarke G, O'Halloran KD, Downer EJ. Impact of Exercise on Innate Immunity in Multiple Sclerosis Progression and Symptomatology. Front Physiol 2016; 7:194. [PMID: 27313534 PMCID: PMC4889582 DOI: 10.3389/fphys.2016.00194] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/16/2016] [Indexed: 12/21/2022] Open
Abstract
Multiple Sclerosis (MS), an idiopathic progressive immune-mediated neurological disorder of the central nervous system (CNS), is characterized by recurrent episodes of inflammatory demyelination and consequent axonal deterioration. It accounts for functional deterioration and lasting disability among young adults. A body of literature demonstrates that physical activity counteracts fatigue and depression and may improve overall quality of life in MS patients. Furthermore, much data indicates that exercise ameliorates chronic neuroinflammation and its related pathologies by tipping cytokine profiles toward an anti-inflammatory signature. Recent data has focused on the direct impact of exercise training on the innate immune system by targeting toll-like receptors (TLRs), signaling pattern recognition receptors that govern the innate immune response, shedding light on the physiological role of TLRs in health and disease. Indeed, TLRs continue to emerge as players in the neuroinflammatory processes underpinning MS. This review will highlight evidence that physical activity and exercise are potential immunomodulatory therapies, targeting innate signaling mechanism(s) to modulate MS symptom development and progression.
Collapse
Affiliation(s)
- Alison Barry
- Department of Physiology, School of Medicine, University College Cork Cork, Ireland
| | - Owen Cronin
- Department of Medicine, Cork University Hospital Cork, Ireland
| | - Aisling M Ryan
- Department of Neurology, Cork University Hospital Cork, Ireland
| | - Brian Sweeney
- Department of Neurology, Cork University Hospital Cork, Ireland
| | | | | | - Andrew P Allen
- Department of Psychiatry and Neurobehavioral Science, APC Microbiome Institute, University College Cork Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioral Science, APC Microbiome Institute, University College Cork Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, University College Cork Cork, Ireland
| | - Eric J Downer
- Department of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin Dublin, Ireland
| |
Collapse
|
199
|
Freedman MS, Rush CA. Severe, Highly Active, or Aggressive Multiple Sclerosis. Continuum (Minneap Minn) 2016; 22:761-84. [DOI: 10.1212/con.0000000000000331] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
200
|
García Merino A, Ramón Ara Callizo J, Fernández Fernández O, Landete Pascual L, Moral Torres E, Rodríguez-Antigüedad Zarrantz A. Consensus statement on the treatment of multiple sclerosis by the Spanish Society of Neurology in 2016. Neurologia 2016; 32:113-119. [PMID: 27157522 DOI: 10.1016/j.nrl.2016.02.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 02/27/2016] [Indexed: 10/21/2022] Open
Abstract
With the advent of new disease-modifying drugs, the treatment of multiple sclerosis is becoming increasingly complex. Using consensus statements is therefore advisable. The present consensus statement, which was drawn up by the Spanish Society of Neurology's study group for demyelinating diseases, updates previous consensus statements on the disease. The present study lists the medications currently approved for multiple sclerosis and their official indications, and analyses such treatment-related aspects as activity, early treatment, maintenance, follow-up, treatment failure, changes in medication, and special therapeutic situations. This consensus statement includes treatment recommendations for a wide range of demyelinating diseases, from isolated demyelinating syndromes to the different forms of multiple sclerosis, as well as recommendations for initial therapy and changes in drug medication, and additional comments on induction and combined therapy and practical aspects of the use of these drugs.
Collapse
Affiliation(s)
| | | | | | | | - E Moral Torres
- Hospital Moisés Broggi, Sant Joan Despí, Barcelona, España
| | | |
Collapse
|