151
|
Fernández O, García-Merino JA, Arroyo R, Álvarez-Cermeño JC, Izquierdo G, Saiz A, Olascoaga J, Rodríguez-Antigüedad A, Prieto JM, Oreja-Guevara C, Hernández MA, Moral E, Meca J, Montalbán X. Spanish consensus on the use of natalizumab (Tysabri®)-2013. Neurologia 2013; 30:302-14. [PMID: 24360652 DOI: 10.1016/j.nrl.2013.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 10/13/2013] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Natalizumab treatment has been shown to be very efficacious in clinical trials and very effective in clinical practice in patients with relapsing-remitting multiple sclerosis, by reducing relapses, slowing disease progression, and improving magnetic resonance imaging patterns. However, the drug has also been associated with a risk of progressive multifocal leukoencephalopathy (PML). The first consensus statement on natalizumab use, published in 2011, has been updated to include new data on diagnostic procedures, monitoring for patients undergoing treatment, PML management, and other topics of interest including the management of patients discontinuing natalizumab. MATERIAL AND METHODS This updated version followed the method used in the first consensus. A group of Spanish experts in multiple sclerosis (the authors of the present document) reviewed all currently available literature on natalizumab and identified the relevant topics would need updating based on their clinical experience. The initial draft passed through review cycles until the final version was completed. RESULTS AND CONCLUSIONS Studies in clinical practice have demonstrated that changing to natalizumab is more effective than switching between immunomodulators. They favour early treatment with natalizumab rather than using natalizumab in a later stage as a rescue therapy. Although the drug is very effective, its potential adverse effects need to be considered, with particular attention to the patient's likelihood of developing PML. The neurologist should carefully explain the risks and benefits of the treatment, comparing them to the risks of multiple sclerosis in terms the patient can understand. Before treatment is started, laboratory tests and magnetic resonance images should be available to permit proper follow-up. The risk of PML should be stratified as high, medium, or low according to presence or absence of anti-JC virus antibodies, history of immunosuppressive therapy, and treatment duration. Although the presence of anti-JC virus antibodies is a significant finding, it should not be considered an absolute contraindication for natalizumab. This update provides general recommendations, but neurologists must use their clinical expertise to provide personalised follow-up for each patient.
Collapse
Affiliation(s)
- O Fernández
- Instituto de Neurociencias Clínicas, Servicio de Neurología, Hospital Regional Universitario Carlos Haya, Fundación Pública Andaluza para la Investigación de Málaga en Biomedicina y Salud, Málaga, España.
| | - J A García-Merino
- Servicio de Neurología, Hospital Universitario Puerta de Hierro, Madrid, España
| | - R Arroyo
- Servicio de Neurología, Hospital Clínico Universitario San Carlos, Madrid, España
| | - J C Álvarez-Cermeño
- Servicio de Neurología, Hospital Universitario Ramón y Cajal, Madrid, España
| | - G Izquierdo
- Servicio de Neurología, Hospital Universitario Virgen Macarena, Sevilla, España
| | - A Saiz
- Servicio de Neurología, Hospital Clinic, Instituto de Investigaciones Biomédicas August Pi i Sunyer, Barcelona, España
| | - J Olascoaga
- Servicio de Neurología, Hospital Universitario de Donostia, San Sebastián, España
| | | | - J M Prieto
- Servicio de Neurología, Hospital Clínico Universitario Santiago de Compostela, Santiago de Compostela, España
| | - C Oreja-Guevara
- Servicio de Neurología, Hospital Clínico Universitario San Carlos, Madrid, España
| | - M A Hernández
- Servicio de Neurología, Hospital Universitario Ntra. Sra. de la Candelaria, Tenerife, España
| | - E Moral
- Servicio de Neurología, Hospital de Sant Joan Despí Moisès Broggi, Barcelona, España
| | - J Meca
- Servicio de Neurología, Hospital Universitario Virgen de la Arrixaca, Murcia, España
| | - X Montalbán
- Centre d'Esclerosi Múltiple de Catalunya (CEMCAT), Hospital Universitari Vall d'Hebron, Barcelona, España
| |
Collapse
|
152
|
Damal K, Stoker E, Foley JF. Optimizing therapeutics in the management of patients with multiple sclerosis: a review of drug efficacy, dosing, and mechanisms of action. Biologics 2013; 7:247-58. [PMID: 24324326 PMCID: PMC3854923 DOI: 10.2147/btt.s53007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis (MS) is a debilitating neurological disorder that affects nearly 2 million adults, mostly in the prime of their youth. An environmental trigger, such as a viral infection, is hypothesized to initiate the abnormal behavior of host immune cells: to attack and damage the myelin sheath surrounding the neurons of the central nervous system. While several other pathways and disease triggers are still being investigated, it is nonetheless clear that MS is a heterogeneous disease with multifactorial etiologies that works independently or synergistically to initiate the aberrant immune responses to myelin. Although there are still no definitive markers to diagnose the disease or to cure the disease per se, research on management of MS has improved many fold over the past decade. New disease-modifying therapeutics are poised to decrease immune inflammatory responses and consequently decelerate the progression of MS disease activity, reduce the exacerbations of MS symptoms, and stabilize the physical and mental status of individuals. In this review, we describe the mechanism of action, optimal dosing, drug administration, safety, and efficacy of the disease-modifying therapeutics that are currently approved for MS therapy. We also briefly touch upon the new drugs currently under investigation, and discuss the future of MS therapeutics.
Collapse
Affiliation(s)
- Kavitha Damal
- Rocky Mountain Multiple Sclerosis Research Group, Salt Lake City, UT, USA
| | | | | |
Collapse
|
153
|
Morris G, Maes M. Myalgic encephalomyelitis/chronic fatigue syndrome and encephalomyelitis disseminata/multiple sclerosis show remarkable levels of similarity in phenomenology and neuroimmune characteristics. BMC Med 2013; 11:205. [PMID: 24229326 PMCID: PMC3847236 DOI: 10.1186/1741-7015-11-205] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 08/15/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND 'Encephalomyelitis disseminata' (multiple sclerosis) and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) are both classified as diseases of the central nervous system by the World Health Organization. This review aims to compare the phenomenological and neuroimmune characteristics of MS with those of ME/CFS. DISCUSSION There are remarkable phenomenological and neuroimmune overlaps between both disorders. Patients with ME/CFS and MS both experience severe levels of disabling fatigue and a worsening of symptoms following exercise and resort to energy conservation strategies in an attempt to meet the energy demands of day-to-day living. Debilitating autonomic symptoms, diminished cardiac responses to exercise, orthostatic intolerance and postural hypotension are experienced by patients with both illnesses. Both disorders show a relapsing-remitting or progressive course, while infections and psychosocial stress play a large part in worsening of fatigue symptoms. Activated immunoinflammatory, oxidative and nitrosative (O+NS) pathways and autoimmunity occur in both illnesses. The consequences of O+NS damage to self-epitopes is evidenced by the almost bewildering and almost identical array of autoantibodies formed against damaged epitopes seen in both illnesses. Mitochondrial dysfunctions, including lowered levels of ATP, decreased phosphocreatine synthesis and impaired oxidative phosphorylation, are heavily involved in the pathophysiology of both MS and ME/CFS. The findings produced by neuroimaging techniques are quite similar in both illnesses and show decreased cerebral blood flow, atrophy, gray matter reduction, white matter hyperintensities, increased cerebral lactate and choline signaling and lowered acetyl-aspartate levels. SUMMARY This review shows that there are neuroimmune similarities between MS and ME/CFS. This further substantiates the view that ME/CFS is a neuroimmune illness and that patients with MS are immunologically primed to develop symptoms of ME/CFS.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Pembrey, Llanelli, UK
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - Michael Maes
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
- Department of Psychiatry, Deakin University, Geelong, Australia
| |
Collapse
|
154
|
Verhey LH, Signori A, Arnold DL, Bar-Or A, Sadovnick AD, Marrie RA, Banwell B, Sormani MP. Clinical and MRI activity as determinants of sample size for pediatric multiple sclerosis trials. Neurology 2013; 81:1215-21. [PMID: 23966255 DOI: 10.1212/wnl.0b013e3182a6cb9b] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To estimate sample sizes for pediatric multiple sclerosis (MS) trials using new T2 lesion count, annualized relapse rate (ARR), and time to first relapse (TTFR) endpoints. METHODS Poisson and negative binomial models were fit to new T2 lesion and relapse count data, and negative binomial time-to-event and exponential models were fit to TTFR data of 42 children with MS enrolled in a national prospective cohort study. Simulations were performed by resampling from the best-fitting model of new T2 lesion count, number of relapses, or TTFR, under various assumptions of the effect size, trial duration, and model parameters. RESULTS Assuming a 50% reduction in new T2 lesions over 6 months, 90 patients/arm are required, whereas 165 patients/arm are required for a 40% treatment effect. Sample sizes for 2-year trials using relapse-related endpoints are lower than that for 1-year trials. For 2-year trials and a conservative assumption of overdispersion (ϑ), sample sizes range from 70 patients/arm (using ARR) to 105 patients/arm (TTFR) for a 50% reduction in relapses, and 230 patients/arm (ARR) to 365 patients/arm (TTFR) for a 30% relapse reduction. Assuming a less conservative ϑ, 2-year trials using ARR require 45 patients/arm (60 patients/arm for TTFR) for a 50% reduction in relapses and 145 patients/arm (200 patients/arm for TTFR) for a 30% reduction. CONCLUSION Six-month phase II trials using new T2 lesion count as an endpoint are feasible in the pediatric MS population; however, trials powered on ARR or TTFR will need to be 2 years in duration and will require multicentered collaboration.
Collapse
Affiliation(s)
- Leonard H Verhey
- From the Pediatric Demyelinating Disease Program (L.H.V.), Program in Neuroscience & Mental Health, The Hospital for Sick Children, University of Toronto, Canada; Biostatistics Unit (A.S., M.P.S.), Department of Health Sciences, University of Genova, Italy; Department of Neurology & Neurosurgery (D.L.A., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Department of Neurology and Division of Medical Genetics (A.D.S.), University of British Columbia, Vancouver; Departments of Internal Medicine and Community Health Sciences (R.A.M.), Winnipeg Health Sciences Centre, University of Manitoba, Winnipeg, Canada; and Division of Neurology (B.B.), Department of Pediatrics, The Children's Hospital of Philadelphia, University of Pennsylvania
| | | | | | | | | | | | | | | |
Collapse
|
155
|
Jones BC, Nair G, Shea CD, Crainiceanu CM, Cortese ICM, Reich DS. Quantification of multiple-sclerosis-related brain atrophy in two heterogeneous MRI datasets using mixed-effects modeling. NEUROIMAGE-CLINICAL 2013; 3:171-9. [PMID: 24179861 PMCID: PMC3791279 DOI: 10.1016/j.nicl.2013.08.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 07/31/2013] [Accepted: 08/01/2013] [Indexed: 11/24/2022]
Abstract
Brain atrophy, measured by MRI, has been proposed as a useful surrogate marker for disease progression in multiple sclerosis (MS). However, it is conventionally assumed that the accurate quantification of brain atrophy is made difficult, if not impossible, by changes in the parameters of the MRI acquisition, which are almost inevitable over the course of a longitudinal study since MRI technology changes rapidly. This state of affairs can negatively affect clinical trial design and limit the use of historical data. Here, we investigate whether we can coherently estimate brain atrophy rates in a heterogeneous MS sample via linear mixed-effects multivariable regression, incorporating three critical assumptions: (1) using age at time of scanning, rather than time since baseline, as the regressor of interest; (2) scanning individuals with a variety of techniques; and (3) introducing a simple additive correction for major differences in MRI protocol. We fit the model to several measures of brain volume as the outcome in two MS populations: 1123 scans from 195 cases acquired for over approximately 7 years in two natural history protocols (Cohort 1), and 1331 scans from 69 cases seen for over 11 years who were primarily treated with two specific MS disease-modifying therapies (Cohort 2). We compared the mixed-effects model with additive correction for MRI acquisition parameters to a model fit without this correction and performed sample-size calculations to provide an estimate of the number of participants in an MS clinical trial that might be required to see a therapeutic effect of treatment using the approach described here. The results show that without the additive correction for T1-weighted protocol parameters, atrophy was underestimated and subject-specific estimates were more narrowly distributed about the population mean. Ventricular CSF is the most consistently estimated brain volume, with a mean of 2.8%/year increase in Cohort 1 and 4.4%/year increase in Cohort 2. An interesting observation was that gray matter volume decreased and white matter volume remained essentially unchanged in both cohorts, suggesting that changes in ventricular CSF volume are a surrogate for changes in gray matter volume. In conclusion, the mixed-effects modeling framework presented here allows effective use of heterogeneously acquired and historical data in the study of brain atrophy in MS, potentially simplifying the design of future single- and multi-site clinical trials and natural history studies. We model brain atrophy in two heterogeneously acquired MS MRI datasets. Mixed-effects regression effectively adjusts for major MRI differences. Ventricular CSF tracks with gray matter but is more reliably estimated. The method would allow reasonable sample sizes in MS therapeutic trials.
Collapse
Affiliation(s)
- Blake C Jones
- Translational Neuroradiology Unit, National Institute of Neurological Disorders and Stroke, NIH, 10 Center Drive MSC 1400, Building 10 Room 5C103, Bethesda, MD 20892, USA ; Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | | | | | | | | | | |
Collapse
|
156
|
Teixeira MZ. Immunomodulatory drugs (natalizumab), worsening of multiple sclerosis, rebound effect and similitude. HOMEOPATHY 2013; 102:215-24. [DOI: 10.1016/j.homp.2013.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 01/15/2013] [Accepted: 05/02/2013] [Indexed: 12/26/2022]
|
157
|
Rudick RA, Fisher E. Preventing brain atrophy should be the gold standard of effective therapy in MS (after the first year of treatment): Yes. Mult Scler 2013; 19:1003-4. [DOI: 10.1177/1352458513482385] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Richard A Rudick
- Mellen Center for MS Treatment and Research, Cleveland, Ohio, USA
| | - Elizabeth Fisher
- Mellen Center for MS Treatment and Research, Cleveland, Ohio, USA
| |
Collapse
|
158
|
Giorgio A, De Stefano N. Clinical use of brain volumetry. J Magn Reson Imaging 2013; 37:1-14. [PMID: 23255412 DOI: 10.1002/jmri.23671] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 03/12/2012] [Indexed: 12/13/2022] Open
Abstract
Magnetic resonance imaging (MRI)-based brain volumetry is increasingly being used in the clinical setting to assess brain volume changes from structural MR images in a range of neurologic conditions. Measures of brain volumes have been shown to be valid biomarkers of the clinical state and progression by offering high reliability and robust inferences on the underlying disease-related mechanisms. This review critically examines the different scenarios of the application of MRI-based brain volumetry in neurology: 1) supporting disease diagnosis, 2) understanding mechanisms and tracking clinical progression of disease, and 3) monitoring treatment effect. These aspects will be discussed in a wide range of neurologic conditions, with particular emphasis on Alzheimer's disease and multiple sclerosis.
Collapse
Affiliation(s)
- Antonio Giorgio
- Department of Neurological and Behavioral Sciences, University of Siena, Italy
| | | |
Collapse
|
159
|
Gladman M, Cudkowicz M, Zinman L. Enhancing clinical trials in neurodegenerative disorders: lessons from amyotrophic lateral sclerosis. Curr Opin Neurol 2013; 25:735-42. [PMID: 23160423 DOI: 10.1097/wco.0b013e32835a309d] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW This review article is focused on strategies that may enhance clinical trial efficiency in neurodegenerative disorders, as demonstrated within the research field of amyotrophic lateral sclerosis (ALS). RECENT FINDINGS Unravelling ALS pathophysiology will result in an increased number of candidate therapeutics. Recent ALS clinical trials have employed novel study designs that expedite the drug development process and limit sample size, including futility, lead-in, selection, adaptive and sequential designs. The search for sensitive and specific biomarkers in ALS continues to develop, and they are essential in accelerating the drug discovery process. Several candidate cerebrospinal fluid (CSF), neuroimaging and electrophysiological biomarkers have been recently described in ALS, and some have been successfully employed as secondary outcome measures in clinical trials. The advent of web-based technologies has provided a complementary platform to expedite clinical trials, through electronic data capture, teleconferencing and online registries. In addition, the formation of ALS consortia has enhanced collaborative multicentre studies. SUMMARY ALS research studies have employed novel strategies to accelerate the efficiency and pace of drug discovery. The importance of adapting to novel measures that enhance study efficiency is not unique to ALS and can be applied to other neurodegenerative diseases in search of effective treatments.
Collapse
Affiliation(s)
- Matthew Gladman
- Department of Medicine, University of Toronto Medical School, Toronto, Ontario, Canada
| | | | | |
Collapse
|
160
|
|
161
|
Arnold DL, Narayanan S, Antel S. Neuroprotection with glatiramer acetate: evidence from the PreCISe trial. J Neurol 2013; 260:1901-6. [PMID: 23589190 PMCID: PMC3705142 DOI: 10.1007/s00415-013-6903-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 03/20/2013] [Accepted: 03/22/2013] [Indexed: 11/01/2022]
Abstract
The phase III, multicenter, randomized, placebo-controlled PreCISe trial assessed glatiramer acetate (GA) effects in patients with clinically isolated syndromes (CIS) suggestive of multiple sclerosis (MS). To assess the neuroprotective effect of GA in a subset of patients in the PreCISe trial, we used proton magnetic resonance spectroscopy (MRS) to measure N-acetylaspartate (NAA), a marker of neuronal integrity, in a large central volume of brain. Thirty-four CIS patients randomized to GA 20 mg/day (n = 19) SC or placebo (n = 15) were included. Patients who relapsed (developed clinically definite MS [CDMS]) were removed from the substudy. NAA/creatine (NAA/Cr) ratios were compared between GA-treated and placebo-treated patients. Twenty patients with CIS had not converted to CDMS and were still in the double-blind phase of the trial at 12 months of follow-up. Paired changes in NAA/Cr differed significantly in patients treated with GA (+0.14, n = 11) compared with patients receiving placebo (-0.33, n = 9, p = 0.03) at 12 months, consistent with a neuroprotective effect of GA in vivo. Patients with CIS who received GA showed improvement in brain neuroaxonal integrity, as indicated by increased NAA/Cr, relative to comparable patients treated with placebo, who showed a decline in NAA/Cr consistent with findings from natural history studies.
Collapse
Affiliation(s)
- Douglas L Arnold
- NeuroRx Research, 3605 University Street, Montreal, QC, H3A 2B3, Canada.
| | | | | |
Collapse
|
162
|
Abstract
INTRODUCTION Monoclonal antibodies (mAbs) represent an emerging and rapidly growing field of therapy in neuroinflammatory diseases. Adhesion molecule blockade by natalizumab represents the first approved mAb therapy in neurology, approved for therapy of highly active multiple sclerosis (MS). Removal of immune cells by anti-CD52 mAb alemtuzumab or anti-CD20 mAb rituximab are other prime examples with existing positive Phase II and Phase III trials. MS clearly represents the neuroinflammatory disease entity with the largest body of evidence. However, some of these approaches are currently investigated or translated for use in other, rare neuroinflammatory diseases, such as neuromyelitis optica (NMO), inflammatory neuropathies and (neuro)-muscular disorders. AREAS COVERED This review will highlight the most relevant therapeutic approaches involving mAbs in the field of neuroinflammatory diseases as published in peer-reviewed journals and presented on international meetings. EXPERT OPINION There is continuously growing evidence on the therapeutic relevance of mAbs in neuroinflammatory disorders. In MS meanwhile several studies have provided evidence for efficacy: In addition to natalizumab, approved in 2006, several other candidates are under development, the most eminent examples with the most advanced study programs being anti-CD52 alemtuzumab, anti-CD20 principles and anti-CD25 daclizumab. Other intriguing candidates are anti-IL-17 strategies, and interference with the complement pathway, partly also developed for other neuroinflammatory disorders.
Collapse
Affiliation(s)
- Luisa Klotz
- Department of Neurology, Inflammatory Disorders of the Nervous System and Neurooncology, Clinic for Neurology, Albert-Schweitzer-Campus 1, Building A10, 48149 Münster, Germany
| | | |
Collapse
|
163
|
Natalizumab treatment reduces fatigue in multiple sclerosis. Results from the TYNERGY trial; a study in the real life setting. PLoS One 2013; 8:e58643. [PMID: 23555589 PMCID: PMC3605436 DOI: 10.1371/journal.pone.0058643] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 02/05/2013] [Indexed: 01/13/2023] Open
Abstract
Fatigue is a significant symptom in multiple sclerosis (MS) patients. First-generation disease modifying therapies (DMTs) are at best moderately effective to improve fatigue. Observations from small cohorts have indicated that natalizumab, an antibody targeting VLA-4, may reduce MS-related fatigue. The TYNERGY study aimed to further evaluate the effects of natalizumab treatment on MS-related fatigue. In this one-armed clinical trial including 195 MS patients, natalizumab was prescribed in a real-life setting, and a validated questionnaire, the Fatigue Scale for Motor and Cognitive functions (FSMC), was used both before and after 12 months of treatment to evaluate a possible change in the fatigue experienced by the patients. In the treated cohort all measured variables, that is, fatigue score, quality of life, sleepiness, depression, cognition, and disability progression were improved from baseline (all p values<0.0001). Walking speed as measured by the six-minute walk-test also increased at month 12 (p = 0.0016). All patients were aware of the nature of the treatment agent, and of the study outcomes.
Collapse
|
164
|
Morsali D, Bechtold D, Lee W, Chauhdry S, Palchaudhuri U, Hassoon P, Snell DM, Malpass K, Piers T, Pocock J, Roach A, Smith KJ. Safinamide and flecainide protect axons and reduce microglial activation in models of multiple sclerosis. ACTA ACUST UNITED AC 2013; 136:1067-82. [PMID: 23518709 DOI: 10.1093/brain/awt041] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Axonal degeneration is a major cause of permanent disability in the inflammatory demyelinating disease multiple sclerosis, but no therapies are known to be effective in axonal protection. Sodium channel blocking agents can provide effective protection of axons in the white matter in experimental models of multiple sclerosis, but the mechanism of action (directly on axons or indirectly via immune modulation) remains uncertain. Here we have examined the efficacy of two sodium channel blocking agents to protect white matter axons in two forms of experimental autoimmune encephalomyelitis, a common model of multiple sclerosis. Safinamide is currently in phase III development for use in Parkinson's disease based on its inhibition of monoamine oxidase B, but the drug is also a potent state-dependent inhibitor of sodium channels. Safinamide provided significant protection against neurological deficit and axonal degeneration in experimental autoimmune encephalomyelitis, even when administration was delayed until after the onset of neurological deficit. Protection of axons was associated with a significant reduction in the activation of microglia/macrophages within the central nervous system. To clarify which property of safinamide was likely to be involved in the suppression of the innate immune cells, the action of safinamide on microglia/macrophages was compared with that of the classical sodium channel blocking agent, flecainide, which has no recognized monoamine oxidase B activity, and which has previously been shown to protect the white matter in experimental autoimmune encephalomyelitis. Flecainide was also potent in suppressing microglial activation in experimental autoimmune encephalomyelitis. To distinguish whether the suppression of microglia was an indirect consequence of the reduction in axonal damage, or possibly instrumental in the axonal protection, the action of safinamide was examined in separate experiments in vitro. In cultured primary rat microglial cells activated by lipopolysaccharide, safinamide potently suppressed microglial superoxide production and enhanced the production of the anti-oxidant glutathione. The findings show that safinamide is effective in protecting axons from degeneration in experimental autoimmune encephalomyelitis, and that this effect is likely to involve a direct effect on microglia that can result in a less activated phenotype. Together, this work highlights the potential of safinamide as an effective neuroprotective agent in multiple sclerosis, and implicates microglia in the protective mechanism.
Collapse
Affiliation(s)
- Damineh Morsali
- Department of Neuroinflammation, UCL Institute of Neurology, Queen Square, 1 Wakefield Street, London, WC1N 1PJ, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
165
|
Kuenz B, Deisenhammer F, Berger T, Reindl M. Diagnostic biomarkers in multiple sclerosis. ACTA ACUST UNITED AC 2013; 1:225-33. [PMID: 23489309 DOI: 10.1517/17530059.1.2.225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the CNS and comprises a heterogeneous spectrum of disease subtypes. The distinctive variability of clinical presentations, histopathologic and immunologic patterns, as well as neuroradiologic phenotypes in MS poses a diagnostic challenge to the attending physician and claims a more differentiated typing of MS patients by diagnostic biomarkers in order to anticipate the expected disease course and to stratify patients for specifically tailored therapies. In this paper, the major biomarkers presently recommended in the diagnosis of MS are reviewed, including magnetic resonance imaging, the analysis of cerebrospinal fluid parameters and the diagnostic relevance of antibodies to aquaporin-4 water channels and myelin antigens.
Collapse
Affiliation(s)
- Bettina Kuenz
- Innsbruck Medical University, Clinical Department of Neurology, Anichstrasse 35, A-6020 Innsbruck, Austria +43 512/504 24363 ; +43 512/504 24266 ;
| | | | | | | |
Collapse
|
166
|
Early magnetic resonance detection of natalizumab-related progressive multifocal leukoencephalopathy in a patient with multiple sclerosis. Case Rep Radiol 2013; 2013:415873. [PMID: 23555065 PMCID: PMC3608276 DOI: 10.1155/2013/415873] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 02/19/2013] [Indexed: 12/19/2022] Open
Abstract
Diagnosis of progressive multifocal leukoencephalopathy is usually based on the clinical presentation, on the demonstration of the brain lesions at the magnetic resonance imaging examination, and on the detection of the JC virus DNA in the cerebrospinal fluid with high sensitive polymerase chain reaction. The role of magnetic resonance imaging specifically in natalizumab-associated progressive multifocal leukoencephalopathy is strengthening, and it is gaining importance not only as an irreplaceable diagnostic tool but also as a surveillance and risk stratifying tool in treated patients. While other imaging techniques such as computed tomography lack sensitivity and specificity, magnetic resonance performed with morphological and functional sequences offers clinicians the possibility to early identify the stage of the disease and the emergence of an immune reconstitution inflammatory syndrome after natalizumab blood removal plasmapheresis.
Collapse
|
167
|
Haanstra KG, Hofman SO, Lopes Estêvão DM, Blezer ELA, Bauer J, Yang LL, Wyant T, Csizmadia V, 't Hart BA, Fedyk ER. Antagonizing the α4β1 integrin, but not α4β7, inhibits leukocytic infiltration of the central nervous system in rhesus monkey experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2013; 190:1961-73. [PMID: 23365083 DOI: 10.4049/jimmunol.1202490] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The immune system is characterized by the preferential migration of lymphocytes through specific tissues (i.e., tissue tropism). Tissue tropism is mediated, in part, by the α(4) integrins expressed by T lymphocytes. The α(4)β(1) integrin mediates migration of memory T lymphocytes into the CNS, whereas the α(4)β(7) integrin mediates migration preferentially into gastrointestinal tissue. This paradigm was established primarily from investigations in rodents; thus, the objective of this investigation was to determine if blocking the α(4)β(7) integrin exclusively would affect migration of T lymphocytes into the CNS of primates. The effects of the dual α(4)β(1) and α(4)β(7) antagonist natalizumab were compared with those of the α(4)β(7) antagonist vedolizumab on experimental autoimmune encephalomyelitis in the rhesus monkey. Animals received an initial i.v. bolus of placebo, natalizumab (30 mg/kg), or vedolizumab (30 mg/kg) before intracutaneous immunization with recombinant human myelin oligodendrocyte glycoprotein and then Ab once weekly thereafter. Natalizumab prevented CNS inflammation and demyelination significantly (p < 0.05), compared with time-matched placebo control animals, whereas vedolizumab did not inhibit these effects, despite saturating the α(4)β(7) integrin in each animal for the duration of the investigation. These results demonstrate that blocking α(4)β(7) exclusively does not inhibit immune surveillance of the CNS in primates.
Collapse
Affiliation(s)
- Krista G Haanstra
- Biomedical Primate Research Centre, 2280 GH Rijswijk, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Hillert J. In the coming year we should abandon interferons and glatiramer acetate as first line therapy for MS: No. Mult Scler 2013; 19:26-8. [DOI: 10.1177/1352458512470312] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet and University Hospital, Stockholm, Sweden
| |
Collapse
|
169
|
|
170
|
Abstract
The treatment of relapsing remitting multiple sclerosis has witnessed major progress since the first effective disease modifying treatment, ß-interferon, became available in 1993. One of the most remarkable new treatments has been natalizumab. This review describes the evolution of this humanized anti-α4ß1 monoclonal antibody, from preclinical experimental research through proof-of-concept (phase 1/2) and pivotal (phase 3) clinical trials to the now extensive experience of its use in clinical practice. The future potential and challenges of natalizumab and oral therapies with a similar mechanism of action are also discussed.
Collapse
Affiliation(s)
- Jeremy Chataway
- Queen Square Multiple Sclerosis Centre, National Hospital for Neurology & Neurosurgery and University College London Institute of Neurology, Queen Square, London, WC1N 3BG, UK.
| | | |
Collapse
|
171
|
MRI outcomes with cladribine tablets for multiple sclerosis in the CLARITY study. J Neurol 2012; 260:1136-46. [PMID: 23263473 DOI: 10.1007/s00415-012-6775-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 10/30/2012] [Accepted: 11/19/2012] [Indexed: 10/27/2022]
Abstract
We herein provide a comprehensive assessment of magnetic resonance imaging (MRI) outcomes from CLARITY, a 96-week, double-blind study demonstrating significant clinical and MRI improvements in patients with relapsing-remitting multiple sclerosis (RRMS) treated with cladribine tablets. Patients with RRMS were randomized 1:1:1 to annual short-course therapy with cladribine tablets cumulative dose 3.5 or 5.25 mg/kg or placebo. MRI endpoints included mean number of T1 gadolinium-enhancing (Gd+), active T2 and combined unique (CU) lesions/patient/scan. MRI-measured disease activity was significantly reduced in both cladribine tablets groups versus placebo. The proportion of patients with no active lesions at study end was: T1 Gd+ lesions: 86.8 and 91.0 versus 48.3 % (p < 0.001); active T2 lesions: 61.7 and 62.5 versus 28.4 % (p < 0.001); CU lesions: 59.6 and 60.7 versus 26.1 % (p < 0.001). Clinically meaningful and significant reductions in active lesion counts and increases in proportions of active lesion-free patients were achieved consistently in cladribine tablet groups when data were stratified by baseline disease characteristics. For example, the percentage of patients who remained lesion-free over the study was significantly greater in cladribine tablet groups than in the placebo group for all lesion types regardless of relapse category at baseline (p < 0.001 for all analyses of patients with ≤1 or 2 relapses; p ≤ 0.022 for analyses of patients with ≥3 relapses). MRI-measured disease activity was greatly reduced by both doses of cladribine tablets, with consistent effect across clinically relevant patient populations. These findings add to our scientific understanding of the neurological impact of this therapeutic modality in patients with RRMS.
Collapse
|
172
|
Jalilian B, Einarsson HB, Vorup-Jensen T. Glatiramer acetate in treatment of multiple sclerosis: a toolbox of random co-polymers for targeting inflammatory mechanisms of both the innate and adaptive immune system? Int J Mol Sci 2012; 13:14579-605. [PMID: 23203082 PMCID: PMC3509598 DOI: 10.3390/ijms131114579] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 10/23/2012] [Accepted: 11/05/2012] [Indexed: 01/17/2023] Open
Abstract
Multiple sclerosis is a disease of the central nervous system, resulting in the demyelination of neurons, causing mild to severe symptoms. Several anti-inflammatory treatments now play a significant role in ameliorating the disease. Glatiramer acetate (GA) is a formulation of random polypeptide copolymers for the treatment of relapsing-remitting MS by limiting the frequency of attacks. While evidence suggests the influence of GA on inflammatory responses, the targeted molecular mechanisms remain poorly understood. Here, we review the multiple pharmacological modes-of-actions of glatiramer acetate in treatment of multiple sclerosis. We discuss in particular a newly discovered interaction between the leukocyte-expressed integrin α(M)β(2) (also called Mac-1, complement receptor 3, or CD11b/CD18) and perspectives on the GA co-polymers as an influence on the function of the innate immune system.
Collapse
Affiliation(s)
- Babak Jalilian
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, Building 1242, DK-8000, Aarhus C, Denmark; E-Mails: (B.J.); (H.B.E.)
| | - Halldór Bjarki Einarsson
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, Building 1242, DK-8000, Aarhus C, Denmark; E-Mails: (B.J.); (H.B.E.)
| | - Thomas Vorup-Jensen
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, Building 1242, DK-8000, Aarhus C, Denmark; E-Mails: (B.J.); (H.B.E.)
| |
Collapse
|
173
|
Vigeveno RM, Wiebenga OT, Wattjes MP, Geurts JJG, Barkhof F. Shifting imaging targets in multiple sclerosis: from inflammation to neurodegeneration. J Magn Reson Imaging 2012; 36:1-19. [PMID: 22696123 DOI: 10.1002/jmri.23578] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Classically multiple sclerosis (MS) has been regarded as an auto-immune disease of the white matter in the central nervous system leading to severe disability over the course of several decades. Current therapeutic strategies in MS are mostly based on either immune suppression or immune modulation. Although effective in decreasing relapse frequency and severity as well as delaying disease progression, MS pathology ensues nonetheless. In the last decade it became evident that gray matter pathology plays an important role in disease progression and helps explaining certain aspects of MS-related disability such as cognitive decline. Conventional MRI outcome measures commonly used in clinical trials are sufficient to demonstrate an anti-inflammatory drug-effect but lack pathological specificity and are poor to moderate predictors of disability. In this article, we review new insights in gray matter pathology and functional reorganization in MS and how these novel fields in MS research may validate and establish new MRI outcome measures, aid in the development of new therapeutic strategies for neuroprotection and neurorepair, and may lead to development of novel predictive measures of disability and disease progression in MS.
Collapse
Affiliation(s)
- René M Vigeveno
- VU University Medical Center, Department of Radiology, Amsterdam, the Netherlands
| | | | | | | | | |
Collapse
|
174
|
Magraner M, Coret F, Casanova B. The relationship between inflammatory activity and brain atrophy in natalizumab treated patients. Eur J Radiol 2012; 81:3485-90. [DOI: 10.1016/j.ejrad.2012.01.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Revised: 01/25/2012] [Accepted: 01/30/2012] [Indexed: 10/28/2022]
|
175
|
Taheri S, Rosenberg GA, Ford C. Quantification of blood-to-brain transfer rate in multiple sclerosis. Mult Scler Relat Disord 2012; 2:124-32. [PMID: 25877634 DOI: 10.1016/j.msard.2012.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 07/30/2012] [Accepted: 09/05/2012] [Indexed: 01/05/2023]
Abstract
Blood-brain barrier (BBB) disruption visualized in lesions by MRI is a major biomarker of disease activity in multiple sclerosis (MS). However, in MS, destruction occurs to a variable extent in lesions as well as in gray matter (GM) and in the normal appearing white matter (NAWM). A method to quantify the BBB disruption in lesions as well as in non-lesion areas would be useful for assessment of MS progression and treatments. The objective of this study was to quantify the BBB transfer rate (Ki) in WM lesions, in the NAWM, and in the full-brain of MS patients. Thirteen MS patients with active lesions and 10 healthy controls with age and gender matching were recruited for full-brain and WM Ki studies. Dynamic contrast-enhanced MRI (DCEMRI) scans were conducted using T1 mapping with partial inversion recovery (TAPIR), a fast T1 mapping technique, following administration of a quarter-dose of the contrast agent Gadolinium-DTPA (Gd-DTPA). The Patlak modeling technique was used to derive a voxel-based map of Ki. In all patients contrast-enhanced lesions, quantified by Ki maps, were observed. Compared with controls, patients with MS exhibited an increase in mean Ki of the full-brain (P-value<0.05) but no significant difference in mean Ki of NAWM. The identified increase in full-brain Ki of MS patients suggests a global vascular involvement associated with MS disease. The lack of observed significant decrease in Ki in NAWM suggests lower involvement of WM vasculature than full-brain vasculature in MS. Ki maps constructed from time series data acquired by DCEMRI provide additional information about BBB that could be used for evaluation of vascular involvement in MS and monitoring treatment effectiveness.
Collapse
Affiliation(s)
- Saeid Taheri
- Department of Radiology and Radiological Sciences, MSC 323, Medical University of South Carolina, Charleston, SC 29425-3230, United States.
| | - Gary A Rosenberg
- Department of Neurology, Departments of Neurosciences, and Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87107, United States
| | - Corey Ford
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87107, United States
| |
Collapse
|
176
|
Ghosh N, Chaki R, Mandal SC. Inhibition of selective adhesion molecules in treatment of inflammatory bowel disease. Int Rev Immunol 2012; 31:410-27. [PMID: 23083349 DOI: 10.3109/08830185.2012.690794] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lymphocyte infiltration into the intestinal tract in inflammatory bowel disease (IBD) is mediated by interaction between α4 integrin and its specific ligands. Development of monoclonal antibodies against α4 integrin allowed targeting of lymphocyte trafficking into the intestine as a novel therapeutic intervention. Natalizumab, vedolizumab, alicaforsen AJM300, rhuMAb β7, CCX282-B, and PF-00547,659 are few of monoclonal antibodies that have shown high promise in trials with the potential for more attractive benefit:risk ratio than currently available therapies. In this review, an attempt is made to underline the therapeutic potential and the safety of anti-adhesion molecule treatment in IBD.
Collapse
Affiliation(s)
- Nilanjan Ghosh
- Dr. B.C. Roy College of Pharmacy and Allied Health Sciences, Durgapur, India. bhu
| | | | | |
Collapse
|
177
|
Bomprezzi R, Okuda DT, Alderazi YJ, Stüve O, Frohman EM. From injection therapies to natalizumab: views on the treatment of multiple sclerosis. Ther Adv Neurol Disord 2012; 5:97-104. [PMID: 22435074 DOI: 10.1177/1756285611431289] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Discoveries of the mechanisms that underlie the pathogenesis of multiple sclerosis have been acquired at an impressive rate over the last few decades and, as a consequence, a growing number of treatments are becoming available for this disease. This review first analyzes the experience from the early stages of the disease-modifying therapies, then, expanding on the concept of early treatment for improved outcomes, it focuses on natalizumab and its major complication, progressive multifocal leukoencephalopathy. We offer views on the risks associated with the use of natalizumab by underscoring the importance of the JC virus serology and by providing preliminary data on our experience with the extended interval dosing of natalizumab. This approach, which advocates individualized treatment plans, raises the question of the minimum effective natalizumab dose. Extended interval dosing suggests efficacy can be maintained while providing advantages of costs and convenience over regular monthly dosing. More data examining this strategy are necessary.
Collapse
|
178
|
Zivadinov R, Dwyer MG, Hussein S, Carl E, Kennedy C, Andrews M, Hojnacki D, Heininen-Brown M, Willis L, Cherneva M, Bergsland N, Weinstock-Guttman B. Voxel-wise magnetization transfer imaging study of effects of natalizumab and IFNβ-1a in multiple sclerosis. Mult Scler 2012; 18:1125-1134. [DOI: 10.1177/1352458511433304] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Objective: To determine the effects of intravenous natalizumab and intramuscular interferon beta-1a (IFNβ-1a) on the volume of white-matter (WM) lesions and normal appearing brain tissue (NABT) undergoing voxel-wise (VW) increases in magnetization transfer ratio (MTR) suggestive of remyelination in patients with relapsing multiple sclerosis. Methods: This prospective, open-label, single-blinded study enrolled patients with relapsing–remitting multiple sclerosis (RRMS) and relapsing secondary progressive multiple sclerosis (RSPMS) as well as a group of age/sex-matched healthy controls ( n=22). Patients with multiple sclerosis were assigned to receive natalizumab monotherapy ( n=77; RRMS/RSPMS) or intramuscular IFNβ-1a ( n=26) as either monotherapy (RRMS) or combined with pulsed i.v. methylprednisolone, as needed (RSPMS). The primary endpoint was the two-year change in volume of NABT VWMTR, by quantifying the number of voxels that increased (suggesting remyelination) or decreased (suggesting demyelination) in their MTR value. Results: The volume of tissue undergoing increases in VWMTR was significantly larger in natalizumab compared with IFNβ-1a-treated patients (year 1: p=0.001 in NABT and p<0.006 in WM lesions; year 2: p=0.008 in NABT) and compared with healthy control subjects (year 1: p=0.05 and year 2: p=0.007 in NABT). The larger volume within NABT undergoing decreases in VWMTR was detected in multiple sclerosis patients compared with healthy controls ( p<0.001), and in the IFNβ-1a group compared with the natalizumab group (year 1: p=0.05; year 2: p=0.002). One patient on natalizumab died from progressive multifocal leukoencephalopathy eight months after completing the study. Conclusion: Natalizumab may promote remyelination and stabilize demyelination in lesions and NABT in relapsing multiple sclerosis, compared with intramuscular IFNβ-1a.
Collapse
Affiliation(s)
- R Zivadinov
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, USA
- The Jacobs Neurological Institute, Department of Neurology, University at Buffalo, State University of New York, Buffalo, USA
| | - MG Dwyer
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, USA
| | - S Hussein
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, USA
| | - E Carl
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, USA
| | - C Kennedy
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, USA
| | - M Andrews
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, USA
| | - D Hojnacki
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, USA
| | - M Heininen-Brown
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, USA
| | - L Willis
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, USA
| | - M Cherneva
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, USA
| | - N Bergsland
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, USA
| | - B Weinstock-Guttman
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, USA
- The Jacobs Neurological Institute, Department of Neurology, University at Buffalo, State University of New York, Buffalo, USA
| |
Collapse
|
179
|
Riley C, Azevedo C, Bailey M, Pelletier D. Clinical applications of imaging disease burden in multiple sclerosis: MRI and advanced imaging techniques. Expert Rev Neurother 2012; 12:323-33. [PMID: 22364331 DOI: 10.1586/ern.11.196] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This review will address the critical role of radiographic techniques in monitoring multiple sclerosis disease course and response to therapeutic interventions using conventional imaging. We propose an algorithm of obtaining a contrast-enhanced brain MRI 6 months after starting a disease-modifying therapy, and considering a gadolinium-enhancing lesion on that scan to indicate suboptimal response to therapy. New or enlarging T2 lesions should be followed on scans at 6-month intervals to assess for change, and the presence of one or more enhancing lesions on a 6- or 12-month scan, or two or more new or enlarging T2 lesions on a 12-month scan should prompt consideration of therapy change. New techniques such as PET imaging, magnetic resonance spectroscopy, magnetic resonance relaxometry, iron-sensitive imaging and perfusion MRI will also be overviewed, with their potential roles in monitoring disease course and activity.
Collapse
Affiliation(s)
- Claire Riley
- Yale University School of Medicine, Yale Multiple Sclerosis Center, 40 Temple St LL, New Haven, CT 06510, USA.
| | | | | | | |
Collapse
|
180
|
Rinaldi F, Calabrese M, Seppi D, Puthenparampil M, Perini P, Gallo P. Natalizumab strongly suppresses cortical pathology in relapsing-remitting multiple sclerosis. Mult Scler 2012; 18:1760-7. [PMID: 22570359 DOI: 10.1177/1352458512447704] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Since cortical pathology has been indicated to play a relevant role in the physical and cognitive disability of multiple sclerosis (MS) patients, this study aims to analyze the efficacy of natalizumab in slowing down its progression. METHODS A total of 120 relapsing-remitting MS patients completed a 2-year prospective study: 35 received natalizumab, 50 received interferon beta-1a or glatiramer acetate (immunomodulatory agents - IMA) and 35 remained untreated. Forty healthy subjects constituted the reference population. Clinical and magnetic resonance imaging (MRI) evaluations (including cortical lesions and atrophy) were performed at baseline and after 2 years. RESULTS Natalizumab significantly reduced accumulation of new cortical lesions (0.2±0.6,range 0-3) compared to immunomodulatory agents (1.3±1.1 togli spazio, range 1-6, p=0.001) and no treatment (2.9±1.5, range 1-8, p<0.001). The percentage of patients with new cortical lesions was also lower in natalizumab-treated patients (20%) compared to IMA-treated and untreated patients (68.0% and 74.2%; p<0.001 for both comparisons). Furthermore, the progression of cortical atrophy was significantly reduced by natalizumab (% change=1.7%) compared to IMA (3.7%, p=0.003) and no therapy (4.6%, p<0.001). Finally, a greater percentage (51.4%) of natalizumab-treated patients remained disease-free (no clinical or MRI evidence of disease activity or progression) compared to IMA-treated (18%, p=0.001) and untreated patients (5.7%, p<0.001). CONCLUSIONS Natalizumab treatment significantly decreases cortical lesion accumulation and cortical atrophy progression in severe relapsing-remitting MS. While supporting the inflammatory origin of cortical lesions, our results highlight the significant impact of natalizumab on cortical pathology.
Collapse
Affiliation(s)
- F Rinaldi
- The Multiple Sclerosis Centre of Veneto Region, Department of Neurology, University Hospital of Padova, Italy
| | | | | | | | | | | |
Collapse
|
181
|
Constantinescu CS, Farooqi N, O'Brien K, Gran B. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol 2012; 164:1079-106. [PMID: 21371012 DOI: 10.1111/j.1476-5381.2011.01302.x] [Citation(s) in RCA: 1076] [Impact Index Per Article: 82.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is the most commonly used experimental model for the human inflammatory demyelinating disease, multiple sclerosis (MS). EAE is a complex condition in which the interaction between a variety of immunopathological and neuropathological mechanisms leads to an approximation of the key pathological features of MS: inflammation, demyelination, axonal loss and gliosis. The counter-regulatory mechanisms of resolution of inflammation and remyelination also occur in EAE, which, therefore can also serve as a model for these processes. Moreover, EAE is often used as a model of cell-mediated organ-specific autoimmune conditions in general. EAE has a complex neuropharmacology, and many of the drugs that are in current or imminent use in MS have been developed, tested or validated on the basis of EAE studies. There is great heterogeneity in the susceptibility to the induction, the method of induction and the response to various immunological or neuropharmacological interventions, many of which are reviewed here. This makes EAE a very versatile system to use in translational neuro- and immunopharmacology, but the model needs to be tailored to the scientific question being asked. While creating difficulties and underscoring the inherent weaknesses of this model of MS in straightforward translation from EAE to the human disease, this variability also creates an opportunity to explore multiple facets of the immune and neural mechanisms of immune-mediated neuroinflammation and demyelination as well as intrinsic protective mechanisms. This allows the eventual development and preclinical testing of a wide range of potential therapeutic interventions.
Collapse
Affiliation(s)
- Cris S Constantinescu
- Division of Clinical Neurology, School of Clinical Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK.
| | | | | | | |
Collapse
|
182
|
Advances in imaging to support the development of novel therapies for multiple sclerosis. Clin Pharmacol Ther 2012; 91:621-34. [PMID: 22398971 DOI: 10.1038/clpt.2011.349] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Multiple sclerosis (MS) is a common neurological disease in North America and Europe. Although most patients develop major locomotor disability over the course of 15-20 years, in approximately one-third of patients the long-term course is favorable, with minimal disability. Although current disease-modifying treatments reduce the relapse rate, their long-term effects are uncertain. MS treatment trials are challenging because of the variable clinical course and typically slow evolution of the disease. Magnetic resonance imaging (MRI) is sensitive in monitoring MS pathology and facilitates evaluation of potential new treatments. MRI measurements of lesion activity have identified new immunomodulatory treatments for preventing relapse. Quantitative measurements of tissue volume and structural integrity, capable of detecting neuroprotection and repair, should facilitate new treatments designed to prevent irreversible disability. Higher-field MR scanners and new positron emission tomography (PET) radioligands are providing new insights into cellular and pathophysiological abnormalities, and should be valuable in future therapeutic trials. Retinal axonal loss measured using optical coherence tomography (OCT) can assess acute neuroprotection in optic neuritis.
Collapse
|
183
|
Gensicke H, Leppert D, Yaldizli Ö, Lindberg RLP, Mehling M, Kappos L, Kuhle J. Monoclonal antibodies and recombinant immunoglobulins for the treatment of multiple sclerosis. CNS Drugs 2012; 26:11-37. [PMID: 22171583 DOI: 10.2165/11596920-000000000-00000] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory and degenerative disease leading to demyelination and axonal damage in the CNS. Autoimmunity plays a central role in MS pathogenesis. Per definition, monoclonal antibodies are recombinant biological compounds with a well defined target, thus carrying the promise of targeting pathogenic cells or molecules with high specificity, avoiding undesired off-target effects. Natalizumab was the first monoclonal antibody to be approved for the treatment of MS. Several other monoclonal antibodies are in development and have demonstrated promising efficacy in phase II studies. They can be categorized according to their mode of action into compounds targeting (i) leukocyte migration into the CNS (natalizumab); (ii) cytolytic antibodies (rituximab, ocrelizumab, ofatumumab, alemtuzumab); or (iii) antibodies and recombinant proteins targeting cytokines and chemokines and their receptors (daclizumab, ustekinumab, atacicept, tabalumab [Ly-2127399], secukinumab [AIN457]). In this review, we discuss the specific molecular targets, clinical efficacy and safety of these compounds and discuss criteria to anticipate the position of monoclonal antibodies in the diversifying armamentarium of MS therapy in the coming years.
Collapse
Affiliation(s)
- Henrik Gensicke
- Neurology, Department of Medicine, University Hospital Basel, Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
184
|
Effect of disease-modifying therapies on brain volume in relapsing–remitting multiple sclerosis: Results of a five-year brain MRI study. J Neurol Sci 2012; 312:7-12. [DOI: 10.1016/j.jns.2011.08.034] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Accepted: 08/24/2011] [Indexed: 11/21/2022]
|
185
|
Barkhof F, Simon JH, Fazekas F, Rovaris M, Kappos L, de Stefano N, Polman CH, Petkau J, Radue EW, Sormani MP, Li DK, O'Connor P, Montalban X, Miller DH, Filippi M. MRI monitoring of immunomodulation in relapse-onset multiple sclerosis trials. Nat Rev Neurol 2011; 8:13-21. [DOI: 10.1038/nrneurol.2011.190] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
186
|
Ziemann U, Wahl M, Hattingen E, Tumani H. Development of biomarkers for multiple sclerosis as a neurodegenerative disorder. Prog Neurobiol 2011; 95:670-85. [DOI: 10.1016/j.pneurobio.2011.04.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 04/07/2011] [Accepted: 04/10/2011] [Indexed: 01/24/2023]
|
187
|
van den Elskamp IJ, Knol DL, Uitdehaag BMJ, Barkhof F. Modeling MR imaging enhancing-lesion volumes in multiple sclerosis: application in clinical trials. AJNR Am J Neuroradiol 2011; 32:2093-7. [PMID: 22051810 DOI: 10.3174/ajnr.a2691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Although the number of enhancing lesions is the typical outcome measure of choice in clinical trials in MS, a potentially more sensitive and statistically more powerful outcome measure is the volume of enhancing lesions. In this study, we assessed the distribution and statistical power of the volume of enhancing brain lesions as an outcome measure by means of their required sample size, and we compared the results with the number of enhancing lesions. MATERIAL AND METHODS First, a literature search was performed to compare the effects of treatment on the number and volume of enhancing lesions. Then, a statistical model was proposed to describe the distribution of the volume of enhancing lesions in 2 datasets of patients with RRMS, and sample sizes for enhancing-lesion volume as primary outcome measure were calculated. RESULTS A mixture of the binomial and Weibull distribution was determined to model enhancing-lesion volumes in patients. Sample size calculations for enhancing-lesion volumes showed that approximately 94 patients per arm would be required to detect a combination of 20% decrease in lesion volume and 20% increase in patients without enhancing lesions, whereas calculations for enhancing-lesion counts showed that approximately 129 patients would be required to detect a 50% decrease. CONCLUSIONS The mixture of the binomial and Weibull distribution is a suitable approach in modeling new enhancing-lesion volumes in MS and yielded feasible sample size estimates for clinical trials, showing lesion volumes to be an advantageous outcome measure compared with lesion counts in terms of statistical power.
Collapse
Affiliation(s)
- I J van den Elskamp
- Department of Radiology, VU University Medical Center, Amsterdam, theNetherlands.
| | | | | | | |
Collapse
|
188
|
Pucci E, Giuliani G, Solari A, Simi S, Minozzi S, Di Pietrantonj C, Galea I. Natalizumab for relapsing remitting multiple sclerosis. Cochrane Database Syst Rev 2011:CD007621. [PMID: 21975773 DOI: 10.1002/14651858.cd007621.pub2] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Natalizumab (NTZ) (Tysabri(®)) is a monoclonal antibody that inhibits leukocyte migration across the blood-brain barrier, thus reducing inflammation in central nervous system, and has been approved worldwide for the treatment of relapsing-remitting multiple sclerosis (RRMS). OBJECTIVES To evaluate the efficacy, tolerability and safety of NTZ in the treatment of patients with RRMS. SEARCH STRATEGY We searched the Cochrane Multiple Sclerosis Group Trials Register, the Cochrane Central Register of Controlled Trials (CENTRAL, The Cochrane Library, 2010, Issue 1), MEDLINE (PubMed) and EMBASE, all up to 19 February 2010, and bibliographies of papers. Handsearching was carried out. Trialists and pharmaceutical companies were contacted. Furthermore, the websites of US Food and Drug Administration (FDA), the European Medicines Evaluation Agency (EMA) and the National Institute for health and Clinical Excellence (NICE) were also checked. SELECTION CRITERIA All double-blind, randomised, controlled trials analysing more than a single infusion of NTZ (dosage > 3 mg/kg intravenous infusion every 4 weeks), also including its use as add-on treatment, versus placebo or other drugs in patients with RRMS. No restrictions on the basis of duration of treatment or length of follow up. DATA COLLECTION AND ANALYSIS Three reviewers independently selected articles which met the inclusion criteria. Disagreements were solved by discussion. Two reviewers independently extracted the data and assessed the methodological quality of each trial. Missing data was sought by contacting principal authors and Biogen Idec, through Biogen-Dompé Italia. MAIN RESULTS Three studies met the inclusion criteria. These included one placebo-controlled trial (942 patients) and two add-on placebo-controlled trials, i.e. one plus glatiramer acetate (110 patients) and the second plus interferon beta-1a (1171 patients).This review assessed the efficacy, tolerability and safety of NTZ in patients with RRMS. Data was conclusive with respect to efficacy and tolerability, but not safety. As far as efficacy is concerned, the results showed statistically significant evidence in favour of NTZ for all the primary outcomes and for the secondary ones where data was available. NTZ reduced the risk of experiencing at least one new exacerbation at 2 years by about 40% and of experiencing progression at 2 years by about 25% as compared to a control group. MRI parameters showed statistical evidence in favour of participants receiving NTZ. Infusion reactions, anxiety, sinus congestion, lower limb swelling, rigors, vaginitis and menstrual disorders were reported as adverse events (AEs) more frequently after NTZ treatment. In this review NTZ was found to be well tolerated over a follow-up period of two years: the number of patients experiencing at least one AE (including severe and serious AEs) during this period did not differ between NTZ-treated patients and controls. Safety concerns have been raised about Progressive Multifocal Leukoencephalopathy (PML). In the trials included in this review, two cases of PML were encountered: one in a patient who had received 29 doses of NTZ and a second fatal case of PML in another patient after 37 doses of NTZ. Our protocol was insufficient to evaluate PML risk as well as other rare and long-term adverse events such as cancers and other opportunistic infections, which are very important issues in considering the risk/benefit ratio of NTZ. AUTHORS' CONCLUSIONS Although one trial did not contribute to efficacy results due to its duration, we found robust evidence in favour of a reduction in relapses and disability at 2 years in RRMS patients treated with NTZ. The drug was well tolerated. There are current significant safety concerns due to reporting of an increasing number of PML cases in patients treated with NTZ. This review was unable to provide an up-to-date systematic assessment of the risk due to the maximum 2 year-duration of the trials included. An independent systematic review of the safety profile of NTZ is warranted. NTZ should be used only by skilled neurologists in MS centres under surveillance programs.All the data in this review came from trials supported by the Pharmaceutical Industry. In agreement with the Cochrane Collaboration policy, this may be considered a potential source of bias.
Collapse
Affiliation(s)
- Eugenio Pucci
- U.O. Neurologia - Ospedale di Macerata, ASUR Marche - Zona Territoriale 9, Via Santa Lucia, 3, Macerata, Italy, 62100
| | | | | | | | | | | | | |
Collapse
|
189
|
|
190
|
Kappos L, Bates D, Edan G, Eraksoy M, Garcia-Merino A, Grigoriadis N, Hartung HP, Havrdová E, Hillert J, Hohlfeld R, Kremenchutzky M, Lyon-Caen O, Miller A, Pozzilli C, Ravnborg M, Saida T, Sindic C, Vass K, Clifford DB, Hauser S, Major EO, O'Connor PW, Weiner HL, Clanet M, Gold R, Hirsch HH, Radü EW, Sørensen PS, King J. Natalizumab treatment for multiple sclerosis: updated recommendations for patient selection and monitoring. Lancet Neurol 2011; 10:745-58. [PMID: 21777829 DOI: 10.1016/s1474-4422(11)70149-1] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Natalizumab, a highly specific α4-integrin antagonist, is approved for treatment of patients with active relapsing-remitting multiple sclerosis (RRMS). It is generally recommended for individuals who have not responded to a currently available first-line disease-modifying therapy or who have very active disease. The expected benefits of natalizumab treatment have to be weighed against risks, especially the rare but serious adverse event of progressive multifocal leukoencephalopathy. In this Review, we revisit and update previous recommendations on natalizumab for treatment of patients with RRMS, based on additional long-term follow-up of clinical studies and post-marketing observations, including appropriate patient selection and management recommendations.
Collapse
|
191
|
Abstract
Owing to its ability to depict the pathologic features of multiple sclerosis (MS) in exquisite detail, conventional magnetic resonance (MR) imaging has become an established tool in the diagnosis of this disease and in monitoring its evolution. MR imaging has been formally included in the diagnostic work-up of patients who present with a clinically isolated syndrome suggestive of MS, and ad hoc diagnostic criteria have been proposed and are updated on a regular basis. In patients with established MS and in those participating in treatment trials, examinations performed with conventional MR pulse sequences provide objective measures to monitor disease activity and progression; however, they have a limited prognostic role. This has driven the application of newer MR imaging technologies, including higher-field-strength MR units, to estimate overall MS burden and mechanisms of recovery in patients at different stages of the disease. These techniques have allowed in vivo assessment of the heterogeneity of MS pathologic features in focal lesions and in normal-appearing tissues. More recently, some of the finer details of MS, including macrophage infiltration and abnormal iron deposition, have become quantifiable with MR imaging. The utility of these modern MR techniques in clinical trial monitoring and in the assessment of the individual patient's response to treatment still need to be evaluated.
Collapse
Affiliation(s)
- Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, Scientific Institute and University Hospital San Raffaele, Via Olgettina 60, 20132 Milan, Italy.
| | | |
Collapse
|
192
|
Fox RJ, Beall E, Bhattacharyya P, Chen JT, Sakaie K. Advanced MRI in multiple sclerosis: current status and future challenges. Neurol Clin 2011; 29:357-80. [PMID: 21439446 DOI: 10.1016/j.ncl.2010.12.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
MRI has rapidly become a leading research tool in the study of multiple sclerosis (MS). Conventional imaging is useful in diagnosis and management of the inflammatory stages of MS but has limitations in describing the degree of tissue injury and cause of progressive disability seen in later stages. Advanced MRI techniques hold promise for filling this void. These imaging tools hold great promise to increase understanding of MS pathogenesis and provide greater insight into the efficacy of new MS therapies.
Collapse
Affiliation(s)
- Robert J Fox
- Mellen Center for Multiple Sclerosis, Neurological Institute, 9500 Euclid Avenue, U-10, Cleveland, OH 44195, USA.
| | | | | | | | | |
Collapse
|
193
|
Abstract
Monoclonal antibodies (MAbs) may have great potential as therapies for autoimmune diseases. Their development as treatments for multiple sclerosis (MS) is promising. Partially effective immunomodulatory therapies have been helpful for many MS patients; however, for patients failing these immunomodulatory treatments, MAbs are an important new treatment option. Currently, MAbs are approved by the US Food and Drug Administration for treatment of many conditions, including autoimmune diseases. This article reviews four MAbs that have been investigated as potential treatments for MS. Of these MAbs, natalizumab is approved for treatment of MS. The other three MAbs (alemtuzumab, rituximab, and daclizumab) are all promising therapies in development for treatment of MS. Adverse effects are relatively mild for these MAbs; however, care in administration and management of these agents is emphasized. Overall, these MAb therapies have great promise in the treatment of MS.
Collapse
|
194
|
Yeh EA, Weinstock-Guttman B. Natalizumab in pediatric multiple sclerosis patients. Ther Adv Neurol Disord 2011; 3:293-9. [PMID: 21179619 DOI: 10.1177/1756285610381526] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Pediatric multiple sclerosis (MS) comprises 2-5% of all cases of MS. Although first-line disease-modifying therapy (DMT) including interferons and glatiramer acetate appear to be well tolerated in this population, recent work has suggested that a growing number of children suffer from disease which is resistant to treatment with these therapies. Natalizumab is a therapy which, although associated with a 1 : 1000 risk for progressive multifocal leukoencephalopathy (PML), has been shown to be well tolerated in the adult population and may lead to disease remission in adults with highly active disease. Reports of use of this therapy in the pediatric population with highly active disease have been published. This paper reviews current experience with the use of natalizumab in the pediatric MS population, with attention to potential risks and possible long-term outcomes in this population.
Collapse
Affiliation(s)
- E Ann Yeh
- Pediatric Multiple Sclerosis and Demyelinating Disorders Center of the Jacobs Neurological Institute, Women and Children's Hospital of Buffalo, and Department of Neurology, State University of New York, Buffalo, NY, USA
| | | |
Collapse
|
195
|
Abstract
Natalizumab is an α4-integrin antagonist approved as monotherapy for patients with relapsing multiple sclerosis (MS), based on demonstrated efficacy in the pivotal AFFIRM study (N = 942). Natalizumab monotherapy reduced risk of disability progression by 42%–54% and annualized relapse rate by 68% during a period of 2 years. Natalizumab was also associated with significant reductions in number of T2-hyperintense, gadolinium-enhancing, and T1-hypointense lesions and in volume of T2-hyperintense lesions (all p < 0.001) on magnetic resonance imaging. Furthermore, natalizumab-treated patients in AFFIRM experienced significant improvements from baseline in the physical and mental components of the Short Form-36 (p ≤ 0.01) and a 35% reduction in risk of clinically significant vision loss (p = 0.008 vs placebo). Natalizumab was well tolerated in phase 3 studies. Common adverse events were generally mild and included headache, fatigue, urinary tract infections, and arthralgia. Serious adverse events were similar between treatment groups. The incidence of serious hypersensitivity reactions associated with natalizumab was <1%. Progressive multifocal leukoencephalopathy was a rare complication of treatment, observed in 2 patients with MS who received natalizumab plus interferon β-1a. The robust clinical benefits of natalizumab, including benefits on patient-reported quality of life, make it an important addition to disease-modifying therapies available to patients with relapsing MS.
Collapse
|
196
|
Natalizumab para la esclerosis múltiple remitente-recurrente. Neurologia 2011; 26:357-68. [DOI: 10.1016/j.nrl.2010.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 06/16/2010] [Indexed: 11/20/2022] Open
|
197
|
|
198
|
Cohen JA, Chun J. Mechanisms of fingolimod's efficacy and adverse effects in multiple sclerosis. Ann Neurol 2011; 69:759-77. [PMID: 21520239 DOI: 10.1002/ana.22426] [Citation(s) in RCA: 293] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Until recently, all approved multiple sclerosis (MS) disease treatments were administered parenterally. Oral fingolimod was approved in September 2010 by the US Food and Drug Administration to reduce relapses and disability progression in relapsing forms of MS. In the clinical trials that led to approval, fingolimod reduced not only acute relapses and magnetic resonance imaging lesion activity but also disability progression and brain volume loss, suggesting preservation of tissue. Fingolimod's mechanism of action in MS is not known with certainty. Its active form, fingolimod-phosphate (fingolimod-P), is a sphingosine 1-phosphate receptor (S1PR) modulator that inhibits egress of lymphocytes from lymph nodes and their recirculation, potentially reducing trafficking of pathogenic cells into the central nervous system (CNS). Fingolimod also readily penetrates the CNS, and fingolimod-P formed in situ may have direct effects on neural cells. Fingolimod potently inhibits the MS animal model, experimental autoimmune encephalomyelitis, but is ineffective in mice with selective deficiency of the S1P₁ S1PR subtype on astrocytes despite normal expression in the immune compartment. These findings suggest that S1PR modulation by fingolimod in both the immune system and CNS, producing a combination of beneficial anti-inflammatory and possibly neuroprotective/reparative effects, may contribute to its efficacy in MS. In clinical trials, fingolimod was generally safe and well tolerated. Its interaction with S1PRs in a variety of tissues largely accounts for the reported adverse effects, which were seen more frequently with doses 2.5 to 10x the approved 0.5 mg dose. Fingolimod's unique mechanism of action distinguishes it from all other currently approved MS therapies.
Collapse
Affiliation(s)
- Jeffrey A Cohen
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | | |
Collapse
|
199
|
Paling D, Golay X, Wheeler-Kingshott C, Kapoor R, Miller D. Energy failure in multiple sclerosis and its investigation using MR techniques. J Neurol 2011; 258:2113-27. [DOI: 10.1007/s00415-011-6117-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 05/18/2011] [Accepted: 05/20/2011] [Indexed: 12/22/2022]
|
200
|
Progressive multifocal leukoencephalopathy and natalizumab. J Neurol 2011; 258:1920-8. [PMID: 21647730 DOI: 10.1007/s00415-011-6116-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/18/2011] [Accepted: 05/20/2011] [Indexed: 01/09/2023]
Abstract
Natalizumab (TYSABRI(®)), a specific α4-integrin antagonist, is approved as a second-line treatment of relapsing-remitting MS (RRMS) patients who fail therapy with interferons or as first-line treatment of patients with highly active relapsing-remitting disease. Since the market introduction of natalizumab as a monotherapy in July of 2006, 111 cases of PML have been reported in natalizumab-treated MS patients as of April 2011. This review focuses on the available data regarding risk stratification for PML under long-term natalizumab therapy, and summarizes the current approach for PML management, as a natalizumab treatment complication is not necessarily associated with a fatal outcome. There is a need for development of surrogate markers that would help to better define the risk of PML in individual patients.
Collapse
|