51
|
Chard DT, Alahmadi AAS, Audoin B, Charalambous T, Enzinger C, Hulst HE, Rocca MA, Rovira À, Sastre-Garriga J, Schoonheim MM, Tijms B, Tur C, Gandini Wheeler-Kingshott CAM, Wink AM, Ciccarelli O, Barkhof F. Mind the gap: from neurons to networks to outcomes in multiple sclerosis. Nat Rev Neurol 2021; 17:173-184. [PMID: 33437067 DOI: 10.1038/s41582-020-00439-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2020] [Indexed: 12/21/2022]
Abstract
MRI studies have provided valuable insights into the structure and function of neural networks, particularly in health and in classical neurodegenerative conditions such as Alzheimer disease. However, such work is also highly relevant in other diseases of the CNS, including multiple sclerosis (MS). In this Review, we consider the effects of MS pathology on brain networks, as assessed using MRI, and how these changes to brain networks translate into clinical impairments. We also discuss how this knowledge can inform the targeting of MS treatments and the potential future directions for research in this area. Studying MS is challenging as its pathology involves neurodegenerative and focal inflammatory elements, both of which could disrupt neural networks. The disruption of white matter tracts in MS is reflected in changes in network efficiency, an increasingly random grey matter network topology, relative cortical disconnection, and both increases and decreases in connectivity centred around hubs such as the thalamus and the default mode network. The results of initial longitudinal studies suggest that these changes evolve rather than simply increase over time and are linked with clinical features. Studies have also identified a potential role for treatments that functionally modify neural networks as opposed to altering their structure.
Collapse
Affiliation(s)
- Declan T Chard
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK. .,National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre, London, UK.
| | - Adnan A S Alahmadi
- Department of Diagnostic Radiology, Faculty of Applied Medical Science, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
| | - Bertrand Audoin
- Aix-Marseille University, CNRS, CRMBM, Marseille, France.,AP-HM, University Hospital Timone, Department of Neurology, Marseille, France
| | - Thalis Charalambous
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Christian Enzinger
- Department of Neurology, Research Unit for Neuronal Repair and Plasticity, Medical University of Graz, Graz, Austria.,Department of Radiology, Division of Neuroradiology, Vascular and Interventional Radiology, Medical University of Graz, Graz, Austria
| | - Hanneke E Hulst
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Maria A Rocca
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Àlex Rovira
- Section of Neuroradiology, Department of Radiology Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jaume Sastre-Garriga
- Servei de Neurologia/Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Menno M Schoonheim
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Betty Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Carmen Tur
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK.,Department of Neurology, Luton and Dunstable University Hospital, Luton, UK
| | - Claudia A M Gandini Wheeler-Kingshott
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK.,Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy.,Brain MRI 3T Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Alle Meije Wink
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Olga Ciccarelli
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK.,National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre, London, UK
| | - Frederik Barkhof
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre, London, UK.,Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Institutes of Neurology and Healthcare Engineering, University College London, London, UK
| | | |
Collapse
|
52
|
Acosta C, Gianinazzi M, Dort T, Armstrong N, Ryder S, Lundqvist T, Ekelund M, Lycke J. Modeling the cost-effectiveness of prolonged-release fampridine for the treatment of walking impairment in patients with multiple sclerosis in Sweden. J Med Econ 2021; 24:770-780. [PMID: 33966549 DOI: 10.1080/13696998.2021.1927746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Aims: To evaluate the cost-effectiveness of adding prolonged-release (PR)-fampridine to best supportive care (BSC) versus BSC alone for the improvement of walking ability in patients with MS.Methods: A cost-utility analysis based on a Markov model was developed to model responders and timed 25-foot walk (T25FW) scores, accumulated costs, and quality-adjusted life-years (QALY) in adults with MS and Expanded Disability Status Scale (EDSS) scores between 4 and 7. The analysis was conducted from a Swedish societal perspective.Results: In the base-case analysis, PR-fampridine plus BSC led to a higher QALY gain than BSC alone. The largest direct cost was professional care provision followed by hospital inpatient stays while the indirect cost was the loss of earnings due to days off work. The incremental cost-effectiveness ratio (ICER) for PR-fampridine plus BSC compared with BSC alone was 57,109 Swedish Kronor (kr)/QALY (€5,607/QALY [1 kr = €0.0981762 on 8 April 2021] and $6,675/QALY [1 kr = $0.116890 on 8 April 2021]). All sensitivity analyses performed resulted in ICERs below 500,000 kr (€49,088 and $58,445).Limitations: Resource use data were not specific to the Swedish market.Conclusions: PR-fampridine represents a cost-effective treatment for MS-related walking impairment in Sweden, due to improvements in patients' quality of life and reduced healthcare resource utilization.
Collapse
Affiliation(s)
- Carlos Acosta
- Value & Market Access, Biogen International GmbH, Baar, Switzerland
| | | | - Thibaut Dort
- Value & Market Access, Biogen International GmbH, Baar, Switzerland
| | | | | | | | - Mats Ekelund
- Market Access, Biogen Sweden AB, Upplands Väsby, Sweden
| | - Jan Lycke
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden
| |
Collapse
|
53
|
Safety, Patient-Reported Well-Being, and Physician-Reported Assessment of Walking Ability in Patients with Multiple Sclerosis for Prolonged-Release Fampridine Treatment in Routine Clinical Practice: Results of the LIBERATE Study. CNS Drugs 2021; 35:1009-1022. [PMID: 34322853 PMCID: PMC8408054 DOI: 10.1007/s40263-021-00840-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2021] [Indexed: 10/26/2022]
Abstract
BACKGROUND Prolonged-release fampridine (PR-FAM) 10-mg tablet twice daily is the only approved pharmacological treatment for improvement of walking ability in adults with multiple sclerosis (MS). LIBERATE assessed the safety/effectiveness of PR-FAM in the real-world. OBJECTIVES The aim of this study was to collect additional safety data, including the incidence rate of seizures and other adverse events (AEs) of interest, from patients with MS taking PR-FAM in routine clinical practice (including patients aged ≥ 65 years and those with pre-existing cardiovascular risk factors). Other objectives included change over time in patient-reported evaluation of physical and psychological impact of MS while taking PR-FAM, and change over time in physician-reported assessment of walking ability in MS patients taking PR-FAM. METHODS Patients with MS newly prescribed PR-FAM were recruited (201 sites, 13 countries). Demographic/safety data were collected at enrolment through 12 months. Physician-rated Clinical Global Impression of Improvement (CGI-I) scores for walking ability, and Multiple Sclerosis Impact Scale-29 (MSIS-29) were assessed. RESULTS Safety analysis included 4646 patients with 3534.8 patient-years of exposure; median (range) age, 52.6 (21-85) years, 87.3% < 65 years, and 65.7% women. Treatment-emergent AEs (TEAEs) were reported in 2448 (52.7%) patients, and serious TEAEs were reported in 279 (6.0%) patients, of whom 37 (< 1%) experienced treatment-emergent serious AEs (TESAEs) considered related to PR-FAM. AEs of special interest (AESI) occurred in 1799 (38.7%) patients, and serious AESI in 128 (2.8%) patients. Seventeen (< 1%) patients experienced actual events of seizure. Overall, 1158 (24.9%) patients discontinued treatment due to lack of efficacy. At 12 months, a greater proportion of patients on-treatment had improvement from baseline in CGI-I for walking ability versus those who discontinued (61% vs. 11%; p < 0.001). MSIS-29 physical impact score improved significantly for patients on-treatment for 12 months versus those who discontinued (mean change, baseline to 12 months: - 9.99 vs. - 0.34 points; p < 0.001). Results were similar for MSIS-29 psychological impact. CONCLUSION No new safety concerns were identified in this real-world study, suggesting that routine risk-minimization measures are effective. CGI-I and MSIS-29 scores after 12 months treatment with PR-FAM treatment show clinical benefits consistent with those previously reported. TRIAL REGISTRATION ClinicalTrials.gov: NCT01480063.
Collapse
|
54
|
Stolyarov ID, Petrov AM, Boyko AN. [Efficacy and safety of Kinezia (fampridine) in the complex therapy of multiple sclerosis]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 120:45-52. [PMID: 33340297 DOI: 10.17116/jnevro202012011145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To analyze the efficacy and safety of fampridine** (Valenta Pharm, Russia) in the complex therapy of multiple sclerosis (MS). MATERIAL AND METHODS One hundred and twenty-six patients with MS were double blind randomized to receive fampridine (n=60) or placebo (n=66). Fampridine was administered in prolonged-release form (film-coated tablets, 10 mg) at a dose of 10 mg (1 tablet) 2 times a day, for 24 weeks. The placebo group was treated in the same way. From the 12th week, all patients in the placebo group were transferred to therapy with fampridine, 10 mg 2 times a day, for another 12 weeks. Concomitant standard therapy for MS was allowed in both groups (concomitant disease-modifying medications and other treatment). The primary outcome in the study was the proportion of patients with reduced t25fw test time (determining walking speed on a 25-foot path) on at least two out of three visits compared to baseline. The mean change in Multiple Sclerosis Functional Composite (MSFC) scores from baseline was assessed at visits 4-7 (8-24 weeks). RESULTS The proportion of patients with reduced t25fw test time compared to the baseline level was 31.7% in the fampridine group, which is higher than in the placebo group - 3.0% (p<0.001). The overall result of the Multiple Sclerosis Functional Composite (MSFC) reflected a gradual improvement in the patient's condition during treatment period. The dynamics of MSFC result relative to the baseline level significantly differed (p<0.05) between the fampridine and placebo groups in favor of the fampridine group during all treatment periods. In the fampridine group, adverse events (AE) associated with disorders of the nervous system were more common: headache, dizziness, and coordination disorders. CONCLUSIONS Fampridine improves walking performance in MS patients. The Russian product fampridine has demonstrated a favorable safety profile.
Collapse
Affiliation(s)
- I D Stolyarov
- N. Bekhtereva Institute of the Human Brain RAS, St.-Petersburg, Russia
| | - A M Petrov
- N. Bekhtereva Institute of the Human Brain RAS, St.-Petersburg, Russia
| | - A N Boyko
- Pirogov Russian National Research Medical University, Moscow, Russia.,Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
| |
Collapse
|
55
|
Donzé C, Papeix C, Lebrun-Frenay C. Urinary tract infections and multiple sclerosis: Recommendations from the French Multiple Sclerosis Society. Rev Neurol (Paris) 2020; 176:804-822. [PMID: 32900473 DOI: 10.1016/j.neurol.2020.02.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVES Establish recommendations for the management of UTIs in MS patients. BACKGROUND Urinary tract infections (UTIs) are common during multiple sclerosis (MS) and are one of the most common comorbidities potentially responsible for deaths from urinary sepsis. METHODS The recommendations attempt to answer three main questions about UTIs and MS. The French Group for Recommendations in MS (France4MS) did a systematic review of articles from PubMed and universities databases (01/1980-12/2019). The RAND/UCLA appropriateness method, which has been developed to synthesize the scientific literature and expert opinions on health care topics, was used for reaching a formal agreement. 26 MS experts worked on the full-text review and a group of 70 multidisciplinary health care specialists validated the final evaluation of summarized evidences. RESULTS UTIs are not associated with an increased risk of relapse and permanent worsening of disability. Only febrile UTIs worsen transient disability through the Uhthoff phenomenon. Some immunosuppressive treatments increase the risk of UTIs in MS patients and require special attention especially in case of hypogammaglobulinemia. Experts recommend to treat UTIs in patients with MS, according to recommendations of the general population. Prevention of recurrent UTIs requires stabilization of the neurogenic bladder. In some cases, weekly oral cycling antibiotics can be proposed after specialist advice. Asymptomatic bacteriuria should not be screened for or treated systematically except in special cases (pregnancy and invasive urological procedures). CONCLUSION Physicians and patients should be aware of the updated recommendations for UTis and MS.
Collapse
Affiliation(s)
- C Donzé
- Faculté de médecine et de maïeutique de Lille, hôpital Saint-Philibert, groupement des hôpitaux de l'institut catholique de Lille, Lomme, France.
| | - C Papeix
- Département de neurologie, CRCSEP, Sorbonne université, hôpital de la Salpêtrière, AP-HP6, Paris 13, France
| | - C Lebrun-Frenay
- URC2A, université Nice Côté-d'Azur, CRCSEP, neurologie hôpital Pasteur 2, CHU de Nice, 30, voie Romaine, 06003 Nice, France
| |
Collapse
|
56
|
HPLC method development for fampridine using Analytical Quality by Design approach. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2020; 70:465-482. [PMID: 32412430 DOI: 10.2478/acph-2020-0036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/20/2019] [Indexed: 01/19/2023]
Abstract
Offering a systematic and multivariate analysis of the analytical procedure, development and validation of HPLC methods using Quality by Design approach are in the limelight of current research trends. A new, experimental design-aided HPLC method for fampridine was developed and preliminarily validated according to current in-force international guidelines for linearity, accuracy, robustness and precision. The method offers a high throughput sample analysis, with an elution time of 2.9 minutes, and signal detection without excipient interference performed at 262 nm. The method proved to be linear between 1-15 µg mL-1 (R2= 0.9996). The mean recovery was found to be 98.7 ± 1.9 % in the tested range of 2.5-7.5 µg mL-1. Low RSD values (< 1 %) were obtained for both model, intra- and inter-day precision. The limit of detection and limit of quantification were 0.24 and 0.78 µg mL-1, resp. The method proved to be applicable for active substance assay in a pharmaceutical dosage form.
Collapse
|
57
|
Beutel T, Dzimiera J, Kapell H, Engelhardt M, Gass A, Schirmer L. Cortical projection neurons as a therapeutic target in multiple sclerosis. Expert Opin Ther Targets 2020; 24:1211-1224. [PMID: 33103501 DOI: 10.1080/14728222.2020.1842358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic inflammatory-demyelinating disease of the central nervous system associated with lesions of the cortical gray matter and subcortical white matter. Recently, cortical lesions have become a major focus of research because cortical pathology and neuronal damage are critical determinants of irreversible clinical progression. Recent transcriptomic studies point toward cell type-specific changes in cortical neurons in MS with a selective vulnerability of excitatory projection neuron subtypes. AREAS COVERED We discuss the cortical mapping and the molecular properties of excitatory projection neurons and their role in MS lesion pathology while placing an emphasis on their subtype-specific transcriptomic changes and levels of vulnerability. We also examine the latest magnetic resonance imaging techniques to study cortical MS pathology as a key tool for monitoring disease progression and treatment efficacy. Finally, we consider possible therapeutic avenues and novel strategies to protect excitatory cortical projection neurons. Literature search methodology: PubMed articles from 2000-2020. EXPERT OPINION Excitatory cortical projection neurons are an emerging therapeutic target in the treatment of progressive MS. Understanding neuron subtype-specific molecular pathologies and their exact spatial mapping will help establish starting points for the development of novel cell type-specific therapies and biomarkers in MS.
Collapse
Affiliation(s)
- Tatjana Beutel
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University , Mannheim, Germany
| | - Julia Dzimiera
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University , Mannheim, Germany
| | - Hannah Kapell
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University , Mannheim, Germany
| | - Maren Engelhardt
- Institute of Neuroanatomy, Medical Faculty Mannheim, MCTN, Heidelberg University , Mannheim, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University , Heidelberg, Germany
| | - Achim Gass
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University , Mannheim, Germany
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University , Mannheim, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University , Heidelberg, Germany
| |
Collapse
|
58
|
De Giglio L, Cortese F, Pennisi EM. Aminopiridines in the treatment of multiple sclerosis and other neurological disorders. Neurodegener Dis Manag 2020; 10:409-423. [PMID: 33054615 DOI: 10.2217/nmt-2020-0018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Symptomatic treatment has a great relevance for the management of patients with neurologic diseases, since it reduces disease burden and improves quality of life. Aminopyridines (APs) are a group of potassium (K+) channel blocking agents that exert their activity both at central nervous system level and on neuromuscular junction. This review describes the use of APs for the symptomatic treatment of neurological conditions. We will describe trials leading to the approval of the extended-release 4-aminopyridine for MS and evidence in support of the use in other neurological diseases.
Collapse
Affiliation(s)
- Laura De Giglio
- Department of Medicine, San Filippo Neri Hospital, Neurology Unit, Rome, Italy
| | - Francesca Cortese
- Department of Medicine, San Filippo Neri Hospital, Neurology Unit, Rome, Italy
| | - Elena Maria Pennisi
- Department of Medicine, San Filippo Neri Hospital, Neurology Unit, Rome, Italy
| |
Collapse
|
59
|
Gunn H, Andrade J, Paul L, Miller L, Creanor S, Stevens K, Green C, Ewings P, Barton A, Berrow M, Vickery J, Marshall B, Zajicek J, Freeman J. A self-management programme to reduce falls and improve safe mobility in people with secondary progressive MS: the BRiMS feasibility RCT. Health Technol Assess 2020; 23:1-166. [PMID: 31217069 DOI: 10.3310/hta23270] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Balance, mobility impairments and falls are common problems for people with multiple sclerosis (MS). Our ongoing research has led to the development of Balance Right in MS (BRiMS), a 13-week home- and group-based exercise and education programme intended to improve balance and encourage safer mobility. OBJECTIVE This feasibility trial aimed to obtain the necessary data and operational experience to finalise the planning of a future definitive multicentre randomised controlled trial. DESIGN Randomised controlled feasibility trial. Participants were block randomised 1 : 1. Researcher-blinded assessments were scheduled at baseline and at 15 and 27 weeks post randomisation. As is appropriate in a feasibility trial, statistical analyses were descriptive rather than involving formal/inferential comparisons. The qualitative elements utilised template analysis as the chosen analytical framework. SETTING Four sites across the UK. PARTICIPANTS Eligibility criteria included having a diagnosis of secondary progressive MS, an Expanded Disability Status Scale (EDSS) score of between ≥ 4.0 and ≤ 7.0 points and a self-report of two or more falls in the preceding 6 months. INTERVENTIONS Intervention - manualised 13-week education and exercise programme (BRiMS) plus usual care. Comparator - usual care alone. MAIN OUTCOME MEASURES Trial feasibility, proposed outcomes for the definitive trial (including impact of MS, mobility, quality of life and falls), feasibility of the BRiMS programme (via process evaluation) and economic data. RESULTS A total of 56 participants (mean age 59.7 years, standard deviation 9.7 years; 66% female; median EDSS score of 6.0 points, interquartile range 6.0-6.5 points) were recruited in 5 months; 30 were block randomised to the intervention group. The demographic and clinical data were broadly comparable at baseline; however, the intervention group scored worse on the majority of baseline outcome measures. Eleven participants (19.6%) withdrew or were lost to follow-up. Worsening of MS-related symptoms unrelated to the trial was the most common reason (n = 5) for withdrawal. Potential primary and secondary outcomes and economic data had completion rates of > 98% for all those assessed. However, the overall return rate for the patient-reported falls diary was 62%. After adjusting for baseline score, the differences between the groups (intervention compared with usual care) at week 27 for the potential primary outcomes were MS Walking Scale (12-item) version 2 -7.7 [95% confidence interval (CI) -17.2 to 1.8], MS Impact Scale (29-item) version 2 (MSIS-29vs2) physical 0.6 (95% CI -7.8 to 9) and MSIS-29vs2 psychological -0.4 (95% CI -9.9 to 9) (negative score indicates improvement). After the removal of one outlier, a total of 715 falls were self-reported over the 27-week trial period, with substantial variation between individuals (range 0-93 falls). Of these 715 falls, 101 (14%) were reported as injurious. Qualitative feedback indicated that trial processes and participant burden were acceptable, and participants highlighted physical and behavioural changes that they perceived to result from undertaking BRiMS. Engagement varied, influenced by a range of condition- and context-related factors. Suggestions to improve the utility and accessibility of BRiMS were highlighted. CONCLUSIONS The results suggest that the trial procedures are feasible and acceptable, and retention, programme engagement and outcome completion rates were sufficient to satisfy the a priori progression criteria. Challenges were experienced in some areas of data collection, such as completion of daily diaries. FUTURE WORK Further development of BRiMS is required to address logistical issues and enhance user-satisfaction and adherence. Following this, a definitive trial to assess the clinical effectiveness and cost-effectiveness of the BRiMS intervention is warranted. TRIAL REGISTRATION Current Controlled Trials ISRCTN13587999. FUNDING This project was funded by the National Institute for Health Research (NIHR) Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 23, No. 27. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Hilary Gunn
- School of Health Professions, Faculty of Health and Human Sciences, Peninsula Allied Health Centre, University of Plymouth, Plymouth, UK
| | - Jackie Andrade
- School of Psychology, Faculty of Health and Human Sciences, University of Plymouth, Plymouth, UK
| | - Lorna Paul
- School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Linda Miller
- Douglas Grant Rehabilitation Unit, Ayrshire Central Hospital, Irvine, UK
| | - Siobhan Creanor
- Peninsula Clinical Trials Unit at Plymouth University (PenCTU), Faculty of Medicine and Dentistry, University of Plymouth, Plymouth, UK.,Medical Statistics Group, Faculty of Medicine and Dentistry, University of Plymouth, Plymouth, UK
| | - Kara Stevens
- Medical Statistics Group, Faculty of Medicine and Dentistry, University of Plymouth, Plymouth, UK
| | - Colin Green
- University of Exeter Medical School, Health Economics Group, University of Exeter, Exeter, UK
| | - Paul Ewings
- National Institute for Health Research (NIHR) Research Design Service (South West), Musgrove Park Hospital, Taunton, UK
| | - Andrew Barton
- National Institute for Health Research (NIHR) Research Design Service, Faculty of Medicine and Dentistry, University of Plymouth, Plymouth, UK
| | - Margie Berrow
- Peninsula Clinical Trials Unit at Plymouth University (PenCTU), Faculty of Medicine and Dentistry, University of Plymouth, Plymouth, UK
| | - Jane Vickery
- Peninsula Clinical Trials Unit at Plymouth University (PenCTU), Faculty of Medicine and Dentistry, University of Plymouth, Plymouth, UK
| | | | - John Zajicek
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Jennifer Freeman
- School of Health Professions, Faculty of Health and Human Sciences, Peninsula Allied Health Centre, University of Plymouth, Plymouth, UK
| |
Collapse
|
60
|
Alshamrani F, Berger W, Alnajashi H, Payne MWC, Morrow SA. Potential of Timed 25-Foot Walk Values in Predicting Maximum Walking Distance in Persons with Multiple Sclerosis. Int J MS Care 2020; 22:215-218. [PMID: 33177957 DOI: 10.7224/1537-2073.2019-052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background Expanded Disability Status Scale (EDSS) scores of 4.0 or greater are determined primarily by maximum walking distance (MWD). Estimation of MWD by persons with multiple sclerosis (MS) is often used due to the impracticality of formally walking a person with MS in a clinic setting. Previous studies have demonstrated discrepancies between estimated and actual MWDs. Whether Timed 25-Foot Walk test (T25FW) values can be used to predict MWD is currently unknown. This study aimed to determine whether T25FW time is predictive of MWD in persons with MS. Methods This study is a post hoc analysis of a previously described prospective cohort study. Persons with MS with an EDSS score of 3.5 to 5.5 were included. The participant's T25FW values and MWD were measured. Results Of the 38 adult participants (mean age, 50.8 years; 27 women [71%]), 24 (63%) had relapsing-remitting MS. The median EDSS score was 4.5 (range, 3.5-5.5). The T25FW times were divided into seven categories (<5.0, 5.0-5.9, 6.0-6.9, 7.0-7.9, 8.0-8.9, 9.0-9.9, and ≥10.0 seconds). The MWDs were divided into corresponding EDSS score categories: ≥500, 300-499, 200-299, 100-199, and ≤99 m. Ordinal logistic regression, when controlled for age, found the T25FW categories to be predictive of EDSS score (χ2 = 17.630, df = 7, P = .014). Conclusions The T25FW value may be used as a surrogate estimate of MWD. Further studies are needed to confirm the reliability of the T25FW in predicting MWD.
Collapse
|
61
|
Soler B, Ramari C, Valet M, Dalgas U, Feys P. Clinical assessment, management, and rehabilitation of walking impairment in MS: an expert review. Expert Rev Neurother 2020; 20:875-886. [PMID: 32729742 DOI: 10.1080/14737175.2020.1801425] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION One of the most common and life-altering consequences of Multiple Sclerosis (MS) is walking impairment. The distance, speed, and Gait pattern functions are components of the International Classification of Functioning, Disability, and Health (ICF) and are also predictors of dependency in terms of daily living activities in patients with MS (pwMS). AREAS COVERED This article provides an overview of walking impairment in pwMS, with focus on the assessment of gait and the rehabilitation approaches. EXPERT OPINION The authors recommend that pwMS undergo gait assessment integrating the ICF perspective using validated clinical outcome measures that cover spatiotemporal gait parameters. Moreover, assessment of walking speed with short walking capacity tests such as the timed 25-foot walk (T25FW) or the 10-m walk test (10 MWT) and tests for walking distance with middle distance tests such as the 2-min walk test (2MWT) and the 6-min walk test (6MWT). This review further highlights strategies that may restore walking function including pharmacological symptomatic treatment and non-pharmacological rehabilitation approaches such as exercise and task-specific training providing an appraisal of mobility targeted therapies to be considered when planning multidisciplinary comprehensive-care of pwMS. Finally, new and novel strategies such as motor imagery and rhythmic auditory stimulation have been developed to improve walking speed and distance in pwMS.
Collapse
Affiliation(s)
- Bernardita Soler
- Neurology Service, Hospital Doctor Sótero Del Río , Santiago, Chile.,Neurology Department, Pontificia Universidad Católica De Chile , Santiago, Chile
| | - Cintia Ramari
- Faculty of Physical Education, University of Brasília , Brasília, Brazil
| | - Maxime Valet
- Cliniques universitaires Saint-Luc, Service de Médecine Physique et Réadaptation , Brussels, Belgium.,Université catholique de Louvain, Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Neuromusculoskeletal lab (NMSK) , Brussels, Belgium
| | - Ulrik Dalgas
- Exercise Biology, Department of Public Health, Aarhus University , Aarhus, Denmark
| | - Peter Feys
- REVAL, Rehabilitation Research Center, Faculty of Rehabilitation Sciences, Hasselt University , Hasselt, Belgium
| |
Collapse
|
62
|
Arpín EC. Efficacy and safety of fampridine for walking disability in multiple sclerosis. Neurodegener Dis Manag 2020; 10:277-287. [PMID: 32762492 DOI: 10.2217/nmt-2020-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Fampridine is the only drug approved for the treatment of walking impairment in multiple sclerosis. Around a third of the patients on treatment obtained an improvement in walking speed during the development phase. The effects are clinically significant, appear soon after the start of the treatment and are long-lasting, but disappear soon after the drug is withdrawn. In the real-world setting, the number of patients with a significant response to the treatment seems to be higher (around 70%). The tolerance is good, with mild to moderate, and transient adverse events. The most commonly reported are insomnia, headache, fatigue, back pain, dizziness, nausea and balance disorders. The main contraindications are a history of seizures, renal impairment and concomitant treatment with OCT2 inhibitors.
Collapse
Affiliation(s)
- Eva Costa Arpín
- Hospital Clínico Universitario, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain
| |
Collapse
|
63
|
Etemadifar M, Saboori M, Chitsaz A, Nouri H, Salari M, Khorvash R, Sheibani Tehrani D, Aghababaee A. The effect of fampridine on the risk of seizure in patients with multiple sclerosis. Mult Scler Relat Disord 2020; 43:102188. [DOI: 10.1016/j.msard.2020.102188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/04/2020] [Accepted: 05/06/2020] [Indexed: 11/27/2022]
|
64
|
Dietrich M, Koska V, Hecker C, Göttle P, Hilla AM, Heskamp A, Lepka K, Issberner A, Hallenberger A, Baksmeier C, Steckel J, Balk L, Knier B, Korn T, Havla J, Martínez-Lapiscina EH, Solà-Valls N, Manogaran P, Olbert ED, Schippling S, Cruz-Herranz A, Yiu H, Button J, Caldito NG, von Gall C, Mausberg AK, Stettner M, Zimmermann HG, Paul F, Brandt AU, Küry P, Goebels N, Aktas O, Berndt C, Saidha S, Green AJ, Calabresi PA, Fischer D, Hartung HP, Albrecht P. Protective effects of 4-aminopyridine in experimental optic neuritis and multiple sclerosis. Brain 2020; 143:1127-1142. [PMID: 32293668 DOI: 10.1093/brain/awaa062] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/08/2019] [Accepted: 01/20/2020] [Indexed: 12/30/2022] Open
Abstract
Chronic disability in multiple sclerosis is linked to neuroaxonal degeneration. 4-aminopyridine (4-AP) is used and licensed as a symptomatic treatment to ameliorate ambulatory disability in multiple sclerosis. The presumed mode of action is via blockade of axonal voltage gated potassium channels, thereby enhancing conduction in demyelinated axons. In this study, we provide evidence that in addition to those symptomatic effects, 4-AP can prevent neuroaxonal loss in the CNS. Using in vivo optical coherence tomography imaging, visual function testing and histologic assessment, we observed a reduction in retinal neurodegeneration with 4-AP in models of experimental optic neuritis and optic nerve crush. These effects were not related to an anti-inflammatory mode of action or a direct impact on retinal ganglion cells. Rather, histology and in vitro experiments indicated 4-AP stabilization of myelin and oligodendrocyte precursor cells associated with increased nuclear translocation of the nuclear factor of activated T cells. In experimental optic neuritis, 4-AP potentiated the effects of immunomodulatory treatment with fingolimod. As extended release 4-AP is already licensed for symptomatic multiple sclerosis treatment, we performed a retrospective, multicentre optical coherence tomography study to longitudinally compare retinal neurodegeneration between 52 patients on continuous 4-AP therapy and 51 matched controls. In line with the experimental data, during concurrent 4-AP therapy, degeneration of the macular retinal nerve fibre layer was reduced over 2 years. These results indicate disease-modifying effects of 4-AP beyond symptomatic therapy and provide support for the design of a prospective clinical study using visual function and retinal structure as outcome parameters.
Collapse
Affiliation(s)
- Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Valeria Koska
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Alexander M Hilla
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Annemarie Heskamp
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Klaudia Lepka
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Andrea Issberner
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Angelika Hallenberger
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| | - Christine Baksmeier
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Julia Steckel
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Lisanne Balk
- Department of Neurology, Amsterdam Neuroscience, MS Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Benjamin Knier
- Department of Experimental Neuroimmunology, Technische Universität München, Munich, Germany
| | - Thomas Korn
- Department of Experimental Neuroimmunology, Technische Universität München, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joachim Havla
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians University, Munich, Germany.,Data Integration for Future Medicine consortium (DIFUTURE), Ludwig-Maximilians University, Munich, Germany
| | - Elena H Martínez-Lapiscina
- Service of Neurology, Hospital Clinic, University of Barcelona, Spain Neuroimmunology Program, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Nuria Solà-Valls
- Service of Neurology, Hospital Clinic, University of Barcelona, Spain Neuroimmunology Program, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Praveena Manogaran
- Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University Hospital Zürich and University of Zürich, Zurich, Switzerland.,Department of Information Technology and Electrical Engineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Elisabeth D Olbert
- Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University Hospital Zürich and University of Zürich, Zurich, Switzerland
| | - Sven Schippling
- Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University Hospital Zürich and University of Zürich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Andrés Cruz-Herranz
- Division of Neuroinflammation and Glial Biology, Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Hao Yiu
- Division of Neuroinflammation and Glial Biology, Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Julia Button
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins Hospital, Baltimore, USA
| | | | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| | - Anne K Mausberg
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Mark Stettner
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Hannah G Zimmermann
- NeuroCure Clinical Research Center and Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Friedemann Paul
- NeuroCure Clinical Research Center and Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Alexander U Brandt
- NeuroCure Clinical Research Center and Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and Max Delbrueck Center for Molecular Medicine, Berlin, Germany.,Department of Neurology, University of California, Irvine, USA
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Norbert Goebels
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Shiv Saidha
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins Hospital, Baltimore, USA
| | - Ari J Green
- Division of Neuroinflammation and Glial Biology, Department of Neurology, University of California San Francisco, San Francisco, USA.,Department of Ophthalmology, University of California San Francisco, San Francisco, USA
| | - Peter A Calabresi
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins Hospital, Baltimore, USA
| | - Dietmar Fischer
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
65
|
Mystery of Expansion: DNA Metabolism and Unstable Repeats. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1241:101-124. [PMID: 32383118 DOI: 10.1007/978-3-030-41283-8_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The mammalian genome mostly contains repeated sequences. Some of these repeats are in the regulatory elements of genes, and their instability, particularly the propensity to change the repeat unit number, is responsible for 36 well-known neurodegenerative human disorders. The mechanism of repeat expansion has been an unsolved question for more than 20 years. There are a few hypotheses describing models of mutation development. Every hypothesis is based on assumptions about unusual secondary structures that violate DNA metabolism processes in the cell. Some models are based on replication errors, and other models are based on mismatch repair or base excision repair errors. Additionally, it has been shown that epigenetic regulation of gene expression can influence the probability and frequency of expansion. In this review, we consider the molecular bases of repeat expansion disorders and discuss possible mechanisms of repeat expansion during cell metabolism.
Collapse
|
66
|
Thompson AK, Sinkjær T. Can Operant Conditioning of EMG-Evoked Responses Help to Target Corticospinal Plasticity for Improving Motor Function in People With Multiple Sclerosis? Front Neurol 2020; 11:552. [PMID: 32765389 PMCID: PMC7381136 DOI: 10.3389/fneur.2020.00552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/15/2020] [Indexed: 11/25/2022] Open
Abstract
Corticospinal pathway and its function are essential in motor control and motor rehabilitation. Multiple sclerosis (MS) causes damage to the brain and descending connections, and often diminishes corticospinal function. In people with MS, neural plasticity is available, although it does not necessarily remain stable over the course of disease progress. Thus, inducing plasticity to the corticospinal pathway so as to improve its function may lead to motor control improvements, which impact one's mobility, health, and wellness. In order to harness plasticity in people with MS, over the past two decades, non-invasive brain stimulation techniques have been examined for addressing common symptoms, such as cognitive deficits, fatigue, and spasticity. While these methods appear promising, when it comes to motor rehabilitation, just inducing plasticity or having a capacity for it does not guarantee generation of better motor functions. Targeting plasticity to a key pathway, such as the corticospinal pathway, could change what limits one's motor control and improve function. One of such neural training methods is operant conditioning of the motor-evoked potential that aims to train the behavior of the corticospinal-motoneuron pathway. Through up-conditioning training, the person learns to produce the rewarded neuronal behavior/state of increased corticospinal excitability, and through iterative training, the rewarded behavior/state becomes one's habitual, daily motor behavior. This minireview introduces operant conditioning approach for people with MS. Guiding beneficial CNS plasticity on top of continuous disease progress may help to prolong the duration of maintained motor function and quality of life in people living with MS.
Collapse
Affiliation(s)
- Aiko K Thompson
- Department of Health Sciences and Research, College of Health Professions, Medical University of South Carolina, Charleston, SC, United States
| | - Thomas Sinkjær
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.,Lundbeck Foundation, Copenhagen, Denmark
| |
Collapse
|
67
|
Strijbis EM, Nij Bijvank JA, Killestein J. 4-aminopyridine is not just a symptomatic therapy, it has a neuroprotective effect - Yes. Mult Scler 2020; 26:1309-1310. [PMID: 32628062 PMCID: PMC7543014 DOI: 10.1177/1352458520923951] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Eva Mm Strijbis
- Department of Neurology, MS Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Jenny A Nij Bijvank
- Department of Neurology, MS Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands/Department of Ophthalmology, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Joep Killestein
- Department of Neurology, MS Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
68
|
Weller D, Lörincz L, Sutter T, Reuter K, Linnebank M, Weller M, Zörner B, Filli L. Fampridine-induced changes in walking kinetics are associated with clinical improvements in patients with multiple sclerosis. J Neurol Sci 2020; 416:116978. [PMID: 32559515 DOI: 10.1016/j.jns.2020.116978] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/07/2020] [Accepted: 06/02/2020] [Indexed: 11/16/2022]
Abstract
Gait dysfunction is common in patients with multiple sclerosis (PwMS). Treatment with prolonged-release fampridine (PR-fampridine) improves walking ability in some PwMS. Associated fampridine-induced changes in the walking pattern are still poorly understood but may provide a better understanding of the mechanisms underlying the beneficial drug effects. 61 PwMS were treated with PR-fampridine in a randomized, monocentric, double-blind and placebo-controlled clinical trial with crossover design (FAMPKIN). Drug-induced improvements in walking speed (Timed-25-Foot Walk; T25FW) and endurance (6-Minute Walk Test; 6MWT) were quantified. In this sub-study of the FAMPKIN trial, fampridine-induced changes in kinetic gait patterns were analyzed by pressure-based foot print analysis during treadmill walking. Vertical ground reaction forces were analyzed during different gait phases. Kinetic data of 44 PwMS was eligible for analysis. During double-blind treatment with PR-fampridine, patients performed significantly better in the T25FW and 6MWT than during placebo treatment (p < 0.0001 for both). At the group level (n = 44), there were no significant changes of gait kinetics under PR-fampridine vs. placebo. However, we found relevant changes of walking kinetics regarding forces during loading, single limb and pre-swing phase in a patient sub-group (n = 8). Interestingly, this sub-group demonstrated superior responsiveness to PR-fampridine in the clinical walking tests compared to those patients without any fampridine-induced changes in kinetics (n = 36). Our results demonstrate fampridine-induced changes in gait kinetics in a sub-group of PwMS. These gait pattern changes were accompanied by improved clinical walking performance under PR-fampridine. These results shed some light on the biomechanical changes in walking patterns underlying enhanced fampridine-induced gait performance.
Collapse
Affiliation(s)
- D Weller
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - L Lörincz
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - T Sutter
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - K Reuter
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - M Linnebank
- Department of Neurology, University Witten/Herdecke and Evangelische Kliniken Gelsenkirchen, Munckelstraße 32, 45879 Gelsenkirchen, Germany
| | - M Weller
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - B Zörner
- Spinal Cord Injury Center, Balgrist University Hospital, Forchstrasse 340, 8008 Zurich, Switzerland
| | - L Filli
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland; Spinal Cord Injury Center, Balgrist University Hospital, Forchstrasse 340, 8008 Zurich, Switzerland; Swiss Center for clinical Movement Analysis (SCMA), Balgrist Campus AG, Lengghalde 5, 8008 Zurich, Switzerland.
| |
Collapse
|
69
|
Munari D, Fonte C, Varalta V, Battistuzzi E, Cassini S, Montagnoli AP, Gandolfi M, Modenese A, Filippetti M, Smania N, Picelli A. Effects of robot-assisted gait training combined with virtual reality on motor and cognitive functions in patients with multiple sclerosis: A pilot, single-blind, randomized controlled trial. Restor Neurol Neurosci 2020; 38:151-164. [DOI: 10.3233/rnn-190974] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Daniele Munari
- Neurorehabilitation Unit, University Hospital of Verona, Verona, Italy
| | - Cristina Fonte
- Neuromotor and Cognitive Rehabilitation Research Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Valentina Varalta
- Neuromotor and Cognitive Rehabilitation Research Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Elisa Battistuzzi
- Neuromotor and Cognitive Rehabilitation Research Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Silvia Cassini
- Neurorehabilitation Unit, University Hospital of Verona, Verona, Italy
| | - Anna Paola Montagnoli
- Neuromotor and Cognitive Rehabilitation Research Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Marialuisa Gandolfi
- Neurorehabilitation Unit, University Hospital of Verona, Verona, Italy
- Neuromotor and Cognitive Rehabilitation Research Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Angela Modenese
- Neuromotor and Cognitive Rehabilitation Research Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Mirko Filippetti
- Neuromotor and Cognitive Rehabilitation Research Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Nicola Smania
- Neurorehabilitation Unit, University Hospital of Verona, Verona, Italy
- Neuromotor and Cognitive Rehabilitation Research Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Alessandro Picelli
- Neurorehabilitation Unit, University Hospital of Verona, Verona, Italy
- Neuromotor and Cognitive Rehabilitation Research Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
70
|
Algahtani HA, Fatani AN, Shirah BH, Algahtani RH. Hashimoto`s Encephalopathy Presenting with Progressive Cerebellar Ataxia. ACTA ACUST UNITED AC 2020; 24:315-319. [PMID: 31872812 PMCID: PMC8015548 DOI: 10.17712/nsj.2019.4.20190016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hashimoto`s encephalopathy is a rare neurological syndrome occurring in patients with autoimmune thyroid disease. The diagnosis of Hashimoto`s encephalopathy is based on the clinical picture with the presence of serum anti-thyroid antibodies regardless of the thyroid disorder. Acquired cerebellar ataxia associated with Hashimoto`s disease is a rare occurrence. In this article, we present a case who had progressive non-familial autoimmune pancerebellar disease in association with an increased level of thyroid peroxidase and thyroglobulin antibodies. The patient was managed aggressively with both intravenous immunoglobulins and plasma exchange, which stopped the progression of the disease and allowed for slow improvement. Early diagnosis of Hashimoto`s encephalopathy with autoimmune cerebellar ataxia and intervention with immunomodulatory therapy are of paramount importance. Close monitoring after steroid therapy is important since some patients with this rare disease might be resistant to steroid therapy and require aggressive immunomodulatory therapy.
Collapse
Affiliation(s)
- Hussein A Algahtani
- King Abdulaziz Medical City, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Kingdom of Saudi Arabia. E-mail:
| | | | | | | |
Collapse
|
71
|
Marion S, Leonid C, Belinda B, Joanne D, Elise H, Leeanne C, Richard M. Effects of modified-release fampridine on upper limb impairment in patients with Multiple Sclerosis. Mult Scler Relat Disord 2020; 40:101971. [PMID: 32062444 DOI: 10.1016/j.msard.2020.101971] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 12/17/2019] [Accepted: 01/27/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND Modified-release 4-aminopyridine (fampridine-MR) is used in the symptomatic treatment of walking disability in patients with multiple sclerosis (MS). Its potential for use in other MS symptoms remains unproven and its mode of action in this context is uncertain. Interest is growing in the use of upper limb outcome measures in clinical trials in patients with Multiple Sclerosis, particularly in advanced or progressive disease. This study tests the following hypotheses: (1) Fampridine-MR improves upper limb function in patients with MS and upper limb impairment. (2) Treatment with fampridine-MR is associated with measurable alterations in objective electrophysiological parameters (evoked potentials and transcranial magnetic stimulation (TMS)) which may predict response to drug treatment. METHODS Study population: patients with MS of any disease subtype, duration and severity who have symptomatic impairment of one or both upper limbs. A healthy control group was included for validation of clinical and electrophysiological measures. Study design: randomised double blind placebo-controlled trial. Treatment details: participants allocated to either fampridine-MR 10 mg bd or placebo of identical appearance for 8 weeks. Primary outcome: performance on 9-hole peg test (9HPT) after 4 weeks. Secondary outcomes: persistence of effect on 9HPT; grip strength; visual acuity and contrast sensitivity; modified fatigue impact scale score; sensory discrimination capacity; visual, somatosensory and motor evoked potentials; resting motor threshold; paired-pulse TMS; peripheral nerve conduction studies. RESULTS 40 patients with MS (60% female, median age 52, median disease duration 13.5 years, median EDSS 6.0) were enrolled. Treatment with fampridine-MR was not associated with any change in upper limb function as measured by the clinical primary or secondary outcomes. Treatment with fampridine-MR was also not associated with any difference in electrophysiological measures of upper limb function. This held true after adjustment for hand dominance, disease duration and severity. Four patients withdrew from the trial because of lack of efficacy or side-effects; all were in the placebo arm. Three patients were admitted to hospital during the study period; one with MS exacerbation (placebo group), one with syncope (drug group) and one with UTI (drug group); otherwise there were no serious adverse events. CONCLUSION Treatment with fampridine-MR was well-tolerated but did not produce clinical benefit in terms of upper limb function, vision or fatigue, nor was there any measurable effect on objective electrophysiological parameters.
Collapse
Affiliation(s)
- Simpson Marion
- Department of Neurology, Austin Hospital, 145 Studley Road, Heidelberg, VIC 3084, Australia; Melbourne Brain Centre, 245 Burgundy Street, Heidelberg, VIC 3084, Australia.
| | - Churilov Leonid
- Melbourne Brain Centre, 245 Burgundy Street, Heidelberg, VIC 3084, Australia.
| | - Bardsley Belinda
- Department of Neurology, Austin Hospital, 145 Studley Road, Heidelberg, VIC 3084, Australia.
| | - Dimovitis Joanne
- Department of Neurology, Austin Hospital, 145 Studley Road, Heidelberg, VIC 3084, Australia.
| | - Heriot Elise
- Department of Neurology, Austin Hospital, 145 Studley Road, Heidelberg, VIC 3084, Australia.
| | - Carey Leeanne
- Department of Neurology, Austin Hospital, 145 Studley Road, Heidelberg, VIC 3084, Australia.
| | - Macdonell Richard
- Department of Neurology, Austin Hospital, 145 Studley Road, Heidelberg, VIC 3084, Australia; Melbourne Brain Centre, 245 Burgundy Street, Heidelberg, VIC 3084, Australia.
| |
Collapse
|
72
|
Rodríguez-Rangel S, Bravin AD, Ramos-Torres KM, Brugarolas P, Sánchez-Rodríguez JE. Structure-activity relationship studies of four novel 4-aminopyridine K + channel blockers. Sci Rep 2020; 10:52. [PMID: 31919372 PMCID: PMC6952366 DOI: 10.1038/s41598-019-56245-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/04/2019] [Indexed: 02/08/2023] Open
Abstract
4-Aminopyridine (4AP) is a specific blocker of voltage-gated potassium channels (KV1 family) clinically approved for the symptomatic treatment of patients with multiple sclerosis (MS). It has recently been shown that [18F]3F4AP, a radiofluorinated analog of 4AP, also binds to KV1 channels and can be used as a PET tracer for the detection of demyelinated lesions in rodent models of MS. Here, we investigate four novel 4AP derivatives containing methyl (-CH3), methoxy (-OCH3) as well as trifluoromethyl (-CF3) in the 2 and 3 position as potential candidates for PET imaging and/or therapy. We characterized the physicochemical properties of these compounds (basicity and lipophilicity) and analyzed their ability to block Shaker K+ channel under different voltage and pH conditions. Our results demonstrate that three of the four derivatives are able to block voltage-gated potassium channels. Specifically, 3-methyl-4-aminopyridine (3Me4AP) was found to be approximately 7-fold more potent than 4AP and 3F4AP; 3-methoxy- and 3-trifluoromethyl-4-aminopyridine (3MeO4AP and 3CF34AP) were found to be about 3- to 4-fold less potent than 4AP; and 2-trifluoromethyl-4-AP (2CF34AP) was found to be about 60-fold less active. These results suggest that these novel derivatives are potential candidates for therapy and imaging.
Collapse
Affiliation(s)
| | - Alyssa D Bravin
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Karla M Ramos-Torres
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Pedro Brugarolas
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| | | |
Collapse
|
73
|
Change in visual acuity and retinal structures following Repository Corticotropin Injection (RCI) therapy in patients with acute demyelinating optic neuritis: Improvement in low contrast visual acuity in both affected and contralateral eyes in a single-armed open-label study. J Neurol Sci 2019; 407:116505. [PMID: 31706456 DOI: 10.1016/j.jns.2019.116505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/22/2019] [Accepted: 09/19/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Current treatments after an episode of optic neuritis have limited success protecting the retinal nerves and restoring visual function. OBJECTIVE To assess the effectiveness of Repository Corticotropin Injection (RCI) after the onset of optic neuritis. METHODS Twenty-four adults were treated with RCI within 2 weeks of symptom onset. Seven exams over 400 days measured low- and high-contrast visual acuity (LCVA and HCVA) and spectral domain optical coherence tomography of the retinal structures. Differences between and among affected and contralateral eyes were assessed using linear mixed models. RESULTS HCVA improved in the affected eye over the study (36.2 letters to 52.5), and LCVA improved in both the affected eye (1.8 letters to 6.8) and the contralateral eye (8.3 letters to 11.7). These functional improvements occurred concurrent to a thinning in the papillomacular bundle and the ganglion cell, inner plexiform, and retinal nerve fiber layers, while the inner nuclear, outer plexiform, outer nuclear, and photoreceptor layers thickened. CONCLUSION The eyes affected by the ON and treated with RCI improved in both LCVA and HCVA, and unexpectedly LCVA improved in the contralateral eye as well. This functional improvement was mirrored by structural changes in the retina. This study lays the groundwork for future studies to explore potential neuro-protective and neuro-restorative effects of RCI.
Collapse
|
74
|
Cortical Excitability Measures May Predict Clinical Response to Fampridine in Patients with Multiple Sclerosis and Gait Impairment. Brain Sci 2019; 9:brainsci9120357. [PMID: 31817319 PMCID: PMC6956091 DOI: 10.3390/brainsci9120357] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/01/2019] [Accepted: 12/03/2019] [Indexed: 02/04/2023] Open
Abstract
Background: Most multiple sclerosis (MS) patients will develop walking limitations during the disease. Sustained-release oral fampridine is the only approved drug that will improve gait in a subset of MS patients. Objectives: (1) Evaluate fampridine cortical excitability effect in MS patients with gait disability. (2) Investigate whether cortical excitability changes can predict the therapeutic response to fampridine. Method: This prospective observational study enrolled 20 adult patients with MS and gait impairment planned to receive fampridine 10 mg twice daily for two consecutive weeks. Exclusion criteria included: Recent relapse (<3 months), modification of disease modifying drugs (<6 months), or Expanded Disability Status Scale (EDSS) score >7. Neurological examination, timed 25-foot walk test (T25wt), EDSS, and cortical excitability studies were performed upon inclusion and 14 days after initiation of fampridine. Results: After treatment, the mean improvement of T25wt (ΔT25wt) was 4.9 s. Significant enhancement of intra-cortical facilitation was observed (139% versus 241%, p = 0.01) following treatment. A positive correlation was found between baseline resting motor threshold (rMT) and both EDSS (r = 0.57; p < 0.01) and ΔT25wt (r = 0.57, p = 0.01). rMT above 52% of the maximal stimulator output was found to be a good predictor of a favorable response to fampridine (accuracy: 75%). Discussion: Fampridine was found to have a significant modulatory effect on the cerebral cortex, demonstrated by an increase in excitatory intracortical processes as unveiled by paired-pulse transcranial magnetic stimulation. rMT could be useful in selecting patients likely to experience a favorable response to fampridine.
Collapse
|
75
|
Walker LAS, Lindsay-Brown AP, Berard JA. Cognitive Fatigability Interventions in Neurological Conditions: A Systematic Review. Neurol Ther 2019; 8:251-271. [PMID: 31586303 PMCID: PMC6858900 DOI: 10.1007/s40120-019-00158-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Although fatigue is a well-studied concept in neurological disease, cognitive fatigability (CF) is less understood. While most studies measure fatigue using subjective self-report, fewer have measured CF objectively. Given the negative impact of CF on quality-of-life, there is a need for targeted interventions. The objective of this review was to determine which procedural, behavioural and pharmacological treatments for objectively measured CF are available to people living with neurological conditions. METHODS In accordance with the PRISMA guidelines, systematic searches for randomized control trials (RCTs), case-controlled studies and case reports/series were conducted across the Ovid Medline, PsycInfo, EMBASE and Cochrane Library databases. English-language articles published between 1980 and February 2019 were considered for eligibility. Included were those that objectively measured CF in individuals with neurological disease/disorder/dysfunction between the ages of 18 and 65 years. Studies were reviewed using a modified Cochrane Data Extraction Template. Risk of bias was assessed using the Cochrane Risk of Bias tool. The review process was facilitated using Covidence software (www.covidence.org). Two authors reviewed articles independently, with a third resolving conflicts regarding article inclusion. RESULTS The search identified 450 records. After duplicates were removed and remaining titles/abstracts were screened for eligibility, 28 full-text articles were assessed, and two studies were included in the qualitative synthesis. Studies were a priori divided into those with pharmacological, procedural or behavioural interventions. Two studies met eligibility criteria; both of these included participants with multiple sclerosis. One study utilized a procedural intervention (i.e. transcranial direct current stimulation), while the other utilized a pharmacological intervention (i.e. fampridine-SR). Studies were evaluated for risk of bias, and evidence from both eligible studies was discussed. CONCLUSION Despite the positive results of the procedural intervention, the paucity of eligible studies and the nascent nature of the field suggests that more studies are required before firm conclusions can be drawn regarding the amenability of CF to treatment. TRIAL REGISTRATION The review was registered with PROSPERO (CRD42019118706).
Collapse
Affiliation(s)
- Lisa A S Walker
- Ottawa Hospital Research Institute, Ottawa, Canada.
- University of Ottawa Brain and Mind Research Institute, Ottawa, Canada.
- Carleton University, Ottawa, Canada.
| | | | | |
Collapse
|
76
|
Valet M, Quoilin M, Lejeune T, Stoquart G, Van Pesch V, El Sankari S, Detrembleur C, Warlop T. Effects of Fampridine in People with Multiple Sclerosis: A Systematic Review and Meta-analysis. CNS Drugs 2019; 33:1087-1099. [PMID: 31612418 DOI: 10.1007/s40263-019-00671-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Prolonged-release (PR) fampridine is a potassium channel blocker used as a symptomatic treatment for walking disability in patients with multiple sclerosis (MS). Its clinical effects in such patients have not been systematically reviewed, and may be more wide-ranging than expected. OBJECTIVES To summarize the evidence on the effects of PR fampridine in patients with MS. METHODS A systematic search of Pubmed, Scopus (including EMBASE), and PsycINFO (completed in 01/2019) was carried out to identify randomized controlled trials (RCT) that compared PR fampridine to placebo. When appropriate, data were pooled using a random-effects model, and standardized mean differences (SMD) were computed. Study quality was assessed using the Downs and Black checklist. PRISMA guidelines were followed. All retrieved functional outcomes were categorized according to the International Classification of Functioning, Disability and Health (ICF). RESULTS A total of 706 articles were screened for inclusion. Twenty RCTs involving 2616 patients met the eligibility criteria. Most studies were of good-to-excellent quality. PR fampridine administration resulted in significant benefits in relation to walking short distances (SMD: 1.23 (95% IC 0.65-1.81)) and perceived walking capacity (0.64 (0.27-1.02)). Its effects on muscle strength and middle-distance walking were not significant (0.53 (- 0.04 to 1.10) and 0.31 (- 0.18 to 0.80), respectively). No effect on higher-level cognitive functions (- 0.07 (- 0.58 to 0.45)) or hand and arm use (0.16 (- 0.33 to 0.64)) was observed. Individual studies reported effects on other outcomes across the ICF domains. CONCLUSIONS There is strong evidence that PR fampridine exerts strong effects on the ability to walk short distances and on perceived walking capacity. Other effects of PR fampridine according to the ICF are possible but still unclear.
Collapse
Affiliation(s)
- Maxime Valet
- Service de Médecine Physique et de Réadaptation, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium. .,Neuromusculoskeletal Lab (NMSK), Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Mounier 53, 1200, Brussels, Belgium.
| | - Mélanie Quoilin
- Service de Médecine Physique et de Réadaptation, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium
| | - Thierry Lejeune
- Service de Médecine Physique et de Réadaptation, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium.,Neuromusculoskeletal Lab (NMSK), Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Mounier 53, 1200, Brussels, Belgium
| | - Gaëtan Stoquart
- Service de Médecine Physique et de Réadaptation, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium.,Neuromusculoskeletal Lab (NMSK), Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Mounier 53, 1200, Brussels, Belgium
| | - Vincent Van Pesch
- Service de Neurologie, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium.,Institute of Neuroscience (IoNS) - Pôle CEMO (Cellular and Molecular), Secteur des Sciences de la Santé, Université catholique de Louvain, Avenue Mounier 53, 1200, Brussels, Belgium
| | - Souraya El Sankari
- Service de Neurologie, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium.,Institute of Neuroscience (IoNS) - Pôle NEUR (Clinical Neuroscience), Secteur des Sciences de la Santé, Université catholique de Louvain, Avenue Mounier 53, 1200, Brussels, Belgium
| | - Christine Detrembleur
- Neuromusculoskeletal Lab (NMSK), Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Mounier 53, 1200, Brussels, Belgium
| | - Thibault Warlop
- Neuromusculoskeletal Lab (NMSK), Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Mounier 53, 1200, Brussels, Belgium.,Service de Neurologie, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium
| |
Collapse
|
77
|
Abstract
Wilhelm Uhthoff, known for his contributions to both neurology and neuro-ophthalmology, was a German ophthalmologist who specialized in neurologic disorders. The eponym "Uhthoff's phenomenon" was first used to describe the reversible, transient blurring of vision in patients with multiple sclerosis during exercise. Subsequently, it was discovered that this neurologic sign not only was triggered by physical exertion but also by other homeostatic disruptions such as hot baths, menstruation, and high external temperatures. Here, we take a look at the life and career of Wilhelm Uhthoff and discuss the basis behind this phenomenon.
Collapse
Affiliation(s)
| | - Mattia Rosso
- Ann Romney Center for Neurologic Disease, Harvard Medical School, Boston, MA, USA
| | - Jonathan D Santoro
- Department of Neurology, Children's Hospital Los Angeles, Los Angeles, CA, USA/Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
78
|
Bozic I, Savic D, Milosevic A, Janjic M, Laketa D, Tesovic K, Bjelobaba I, Jakovljevic M, Nedeljkovic N, Pekovic S, Lavrnja I. The Potassium Channel Kv1.5 Expression Alters During Experimental Autoimmune Encephalomyelitis. Neurochem Res 2019; 44:2733-2745. [PMID: 31624998 DOI: 10.1007/s11064-019-02892-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/23/2019] [Accepted: 10/11/2019] [Indexed: 12/29/2022]
Abstract
Multiple sclerosis (MS) is a chronic, inflammatory, neurodegenerative disease with an autoimmune component. It was suggested that potassium channels, which are involved in crucial biological functions may have a role in different diseases, including MS and its animal model, experimental autoimmune encephalomyelitis (EAE). It was shown that voltage-gated potassium channels Kv1.5 are responsible for fine-tuning in the immune physiology and influence proliferation and differentiation in microglia and astrocytes. Here, we explored the cellular distribution of the Kv1.5 channel, together with its transcript and protein expression in the male rat spinal cord during different stages of EAE. Our results reveal a decrease of Kv1.5 transcript and protein level at the peak of disease, where massive infiltration of myeloid cells occurs, together with reactive astrogliosis and demyelination. Also, we revealed that the presence of this channel is not found in infiltrating macrophages/microglia during EAE. It is interesting to note that Kv1.5 channel is expressed only in resting microglia in the naïve animals. Predominant expression of Kv1.5 channel was found in the astrocytes in all experimental groups, while some vimentin+ cells, resembling macrophages, are devoid of Kv1.5 expression. Our results point to the possible link between Kv1.5 channel and the pathophysiological processes in EAE.
Collapse
Affiliation(s)
- I Bozic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - D Savic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - A Milosevic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - M Janjic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - D Laketa
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - K Tesovic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - I Bjelobaba
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - M Jakovljevic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - N Nedeljkovic
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - S Pekovic
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - I Lavrnja
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
79
|
Srinivasan SR, Shakkottai VG. Moving Towards Therapy in SCA1: Insights from Molecular Mechanisms, Identification of Novel Targets, and Planning for Human Trials. Neurotherapeutics 2019; 16:999-1008. [PMID: 31338702 PMCID: PMC6985354 DOI: 10.1007/s13311-019-00763-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The spinocerebellar ataxias (SCAs) are a group of neurodegenerative disorders inherited in an autosomal dominant fashion. The SCAs result in progressive gait imbalance, incoordination of the limbs, speech changes, and oculomotor dysfunction, among other symptoms. Over the past few decades, significant strides have been made in understanding the pathogenic mechanisms underlying these diseases. Although multiple efforts using a combination of genetics and pharmacology with small molecules have been made towards developing new therapeutics, no FDA approved treatment currently exists. In this review, we focus on SCA1, a common SCA subtype, in which some of the greatest advances have been made in understanding disease biology, and consequently potential therapeutic targets. Understanding of the underlying basic biology and targets of therapy in SCA1 is likely to give insight into treatment strategies in other SCAs. The diversity of the biology in the SCAs, and insight from SCA1 suggests, however, that both shared treatment strategies and specific approaches tailored to treat distinct genetic causes of SCA are likely needed for this group of devastating neurological disorders.
Collapse
Affiliation(s)
| | - Vikram G Shakkottai
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.
- Department of Molecular and Integrative Physiology, University of Michigan, 4009 BSRB, 109 Zina Pitcher Place, Ann Arbor, Michigan, 48109, USA.
| |
Collapse
|
80
|
Study on Dalfampridine in the treatment of Multiple Sclerosis Mobility Disability: A meta-analysis. PLoS One 2019; 14:e0222288. [PMID: 31513613 PMCID: PMC6742383 DOI: 10.1371/journal.pone.0222288] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 08/26/2019] [Indexed: 11/19/2022] Open
Abstract
Objective Systematic Review was used to evaluate the efficacy and safety of Dalfampridine (DAP) in the treatment of Mobility Disability (MS) in patients with Multiple Sclerosis. Methods Clinical randomized controlled studies about DAP and placebo in the treatment of Mobility Disability in patients with Multiple Sclerosis until March 2019 were explored by searching Embase, PubMed, Cochrane, Web of Knowledge, and ClinicalTrials.gov. Literature screening, data extraction, quality assessment, and statistical analysis were performed by using Stata 14.0. Results 10 papers were included in the meta-analysis, and the number of patients was 2100. In conclusion, the application of DAP in clinical can significantly improve the Mobility Disability of patients [OR = 2.73, 95%CI (1.66, 4.50), P<0.001, I2 = 74.1%] and boost the mobility speed of patients in Timing 24 Minute Walk Test (T24FW) [SMD = 3,08, 95%CI(1,58, 4.58), P<0.001, I2 = 98.7%]. There are no significant differences of the incidence of adverse events [RR = 1.06, 95%CI (0.99, 1.14), P = 0.928, I2 = 0.0%] and urinary tract infection [RR = 1.21, 95%CI(0.91, 1.60), P = 0.145, I2 = 37.2%] between the DAP test group (Doses≤10 mg) and the placebo control group, and the incidence of adverse events [RR = 1.14, 95%CI(1.02, 1.28), P = 0.793, I2 = 0.0%] and urinary tract infection[RR = 3.05, 95%CI(1.04, 8.99), P = 0.680, I2 = 0.0%] for the DAP test group (Doses>10 mg) is a litter higher than the placebo control group. Conclusion DAP can effectively improve Mobility Disability in patients with Multiple Sclerosis, which is safe and reliable in specific DAP usage doses.
Collapse
|
81
|
Abstract
The spinocerebellar ataxias (SCAs) comprise more than 40 autosomal dominant neurodegenerative disorders that present principally with progressive ataxia. Within the past few years, studies of pathogenic mechanisms in the SCAs have led to the development of promising therapeutic strategies, especially for SCAs caused by polyglutamine-coding CAG repeats. Nucleotide-based gene-silencing approaches that target the first steps in the pathogenic cascade are one promising approach not only for polyglutamine SCAs but also for the many other SCAs caused by toxic mutant proteins or RNA. For these and other emerging therapeutic strategies, well-coordinated preparation is needed for fruitful clinical trials. To accomplish this goal, investigators from the United States and Europe are now collaborating to share data from their respective SCA cohorts. Increased knowledge of the natural history of SCAs, including of the premanifest and early symptomatic stages of disease, will improve the prospects for success in clinical trials of disease-modifying drugs. In addition, investigators are seeking validated clinical outcome measures that demonstrate responsiveness to changes in SCA populations. Findings suggest that MRI and magnetic resonance spectroscopy biomarkers will provide objective biological readouts of disease activity and progression, but more work is needed to establish disease-specific biomarkers that track target engagement in therapeutic trials. Together, these efforts suggest that the development of successful therapies for one or more SCAs is not far away.
Collapse
|
82
|
De Giglio L, De Luca F, Gurreri F, Ferrante I, Prosperini L, Borriello G, Quartuccio E, Gasperini C, Pozzilli C. Effect of dalfampridine on information processing speed impairment in multiple sclerosis. Neurology 2019; 93:e733-e746. [DOI: 10.1212/wnl.0000000000007970] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 04/30/2019] [Indexed: 02/02/2023] Open
Abstract
ObjectiveTo test a possible benefit of dalfampridine on information processing speed (IPS), a key function for cognitive impairment (CogIm) in multiple sclerosis (MS).MethodsIn this randomized, double-blind, placebo-controlled trial, we included patients with a score on the Symbol Digit Modalities Test (SDMT) under the 10th percentile of the reference value. Patients were randomized in a 2:1 ratio to receive dalfampridine 10 mg or placebo twice daily for 12 weeks. They underwent a comprehensive neuropsychological evaluation at screening (T0), at the end of treatment (T1), and after a 4-week follow-up (T2). The primary endpoint was improvement in SDMT.ResultsOut of 208 patients screened, 120 were randomized to receive either dalfampridine (n = 80) or placebo (n = 40). At T1, the dalfampridine group presented an increase of SDMT scores vs placebo group (mean change 9.9 [95% confidence interval (CI) 8.5–11.4] vs 5.2 [95% CI 2.8–7.6], p = 0.0018; d = 0.60 for raw score; and 0.8 [95% CI 0.6–1] vs 0.3 [95% CI 0.0–0.5], p = 0.0013; d = 0.61 for z scores; by linear mixed model with robust standard error). The improvement was not sustained at T2. A beneficial effect of dalfampridine was observed in the Paced Auditory Serial Addition Test and in cognitive fatigue.ConclusionDalfampridine could be considered as an effective treatment option for IPS impairment in MS.Trial registration2013-002558-64 EU Clinical Trials Register.Classification of evidenceThis study provides Class I evidence that for patients with MS with low scores on the SDMT, dalfampridine improves IPS.
Collapse
|
83
|
Foschi M, Lugaresi A. Evaluating dalfampridine for the treatment of relapsing-remitting multiple sclerosis: does it add to the treatment armamentarium? Expert Opin Pharmacother 2019; 20:1309-1320. [PMID: 31237780 DOI: 10.1080/14656566.2019.1623879] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Multiple sclerosis (MS) is a demyelinating disease, causing axonal damage and disability. Dalfampridine (DAL) is an extended-release formulation of 4-aminopyridine (4AP) and broad-spectrum voltage-dependent potassium channel blocker that is reported to improve motor, visual and cognitive functions. Furthermore, it is presently the only approved drug for walking impairment in MS. Areas covered: Herein, the authors evaluate DAL as a relapsing-remitting MS treatment, reporting and commenting on all aspects of the drug including its chemistry, safety, pharmacokinetics, and cost-effectiveness. A bibliographic search was performed on PubMed using the terms 'dalfampridine OR fampridine OR 4-aminopyridine'. Expert opinion: Evidence from post-marketing studies suggests that DAL, consistent with the effects of 4AP, may not only improve walking speed, but also arm function, fatigue, mood and cognition through restored nerve conduction in central nervous system demyelinated areas. Long-term safety data confirm that the approved dose of 10 mg twice daily is generally well tolerated. However, despite the reported efficacy, the extent of the benefits is limited in real life activities, although significant improvements have been demonstrated in the clinical setting. Patients often complain of side effects (such as cramps and painful paraesthesia) or lack of efficacy. Also, its considerably higher pricing in comparison to 4AP represents an important limitation.
Collapse
Affiliation(s)
- Matteo Foschi
- a Dipartimento di Scienze Biomediche e Neuromotorie , Università di Bologna , Bologna , Italy
| | - Alessandra Lugaresi
- a Dipartimento di Scienze Biomediche e Neuromotorie , Università di Bologna , Bologna , Italy.,b IRCCS Istituto delle Scienze Neurologiche di Bologna , UOSI Riabilitazione Sclerosi Multipla , Bologna , Italy
| |
Collapse
|
84
|
Amifampridine Phosphate (Firdapse) Is Effective in a Confirmatory Phase 3 Clinical Trial in LEMS. J Clin Neuromuscul Dis 2019; 20:111-119. [PMID: 30801481 PMCID: PMC6392213 DOI: 10.1097/cnd.0000000000000239] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Objective: To assess tolerability and efficacy of amifampridine phosphate versus placebo for symptomatic treatment of Lambert–Eaton Myasthenic Syndrome (LEMS). Methods: This phase 3 randomized, double-blind, placebo-controlled withdrawal trial in 26 adults with LEMS compared efficacy of amifampridine phosphate versus placebo over a 4-day period. The primary endpoints were quantitative myasthenia gravis score (QMG) and subject global impression, and the secondary endpoint was Clinical Global Impression–Improvement. The exploratory endpoints were 3TUG (timed up and go) test and QMG limb domain score. All participants had been receiving amifampridine phosphate (30–80 mg/d divided into 3 or 4 doses daily) in an expanded access protocol and had been titrated to the optimal dose and frequency for at least 1 week before randomization into the current study. After completion of assessments after 4 days of double-blind treatment, patients had the option to return to open-label amifampridine phosphate. The efficacy endpoints were mean changes from baseline in the various evaluation parameters. Results: Amifampridine phosphate (n = 13) demonstrated significant benefit in QMG and subject global impression compared with placebo (n = 13) at 4 days. Other measures of efficacy, including Clinical Global Impression–Improvement, 3TUG, and QMG limb domain score were also improved. The most common “adverse events” in the placebo group were muscle weakness (n = 5) and fatigue (n = 4), as expected from withdrawal of amifampridine phosphate, whereas only back pain (n = 1), pain in extremity (n = 1), and headache (n = 1) were reported in amifampridine phosphate group. Conclusions: This phase 3 randomized, double-blind, placebo-controlled withdrawal trial in adults with LEMS provided class I evidence of efficacy of amifampridine phosphate as symptomatic treatment in LEMS.
Collapse
|
85
|
Wulff H, Christophersen P, Colussi P, Chandy KG, Yarov-Yarovoy V. Antibodies and venom peptides: new modalities for ion channels. Nat Rev Drug Discov 2019; 18:339-357. [PMID: 30728472 PMCID: PMC6499689 DOI: 10.1038/s41573-019-0013-8] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Ion channels play fundamental roles in both excitable and non-excitable tissues and therefore constitute attractive drug targets for myriad neurological, cardiovascular and metabolic diseases as well as for cancer and immunomodulation. However, achieving selectivity for specific ion channel subtypes with small-molecule drugs has been challenging, and there currently is a growing trend to target ion channels with biologics. One approach is to improve the pharmacokinetics of existing or novel venom-derived peptides. In parallel, after initial studies with polyclonal antibodies demonstrated the technical feasibility of inhibiting channel function with antibodies, multiple preclinical programmes are now using the full spectrum of available technologies to generate conventional monoclonal and engineered antibodies or nanobodies against extracellular loops of ion channels. After a summary of the current state of ion channel drug discovery, this Review discusses recent developments using the purinergic receptor channel P2X purinoceptor 7 (P2X7), the voltage-gated potassium channel KV1.3 and the voltage-gated sodium channel NaV1.7 as examples of targeting ion channels with biologics.
Collapse
Affiliation(s)
- Heike Wulff
- Department of Pharmacology, University of California Davis, Davis, CA, USA.
| | | | | | - K George Chandy
- Molecular Physiology Laboratory, Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Vladimir Yarov-Yarovoy
- Department of Physiology & Membrane Biology, University of California Davis, Davis, CA, USA
| |
Collapse
|
86
|
Rodriguez-Leal FA, Haase R, Akgün K, Eisele J, Proschmann U, Schultheiss T, Kern R, Ziemssen T. Nonwalking response to fampridine in patients with multiple sclerosis in a real-world setting. Ther Adv Chronic Dis 2019; 10:2040622319835136. [PMID: 31037211 PMCID: PMC6475844 DOI: 10.1177/2040622319835136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/06/2019] [Indexed: 11/15/2022] Open
Abstract
Objectives Mobility impairments constitute a long-term burden in patients with multiple sclerosis (MS). Currently there is evidence that the drug fampridine may improve nonwalking symptoms in MS patients. The main objective of this study is to analyze whether participants showing a beneficial walking response to fampridine, also show a positive response in nonwalking assessments in a real-world clinical setting. Methods Subjects enrolled were part of a study analyzing gait parameters, for which response to treatment with fampridine was monitored after a period of 2 weeks. Neurologists then decided whether patients were responders to fampridine (RF) according to their global impression of patients' gait improvement. As nonwalking outcomes, we included the nine-hole peg test (9-HPT), the EuroQoL five dimensions questionnaire (EQ-5D) for quality of life, The Würzburger Fatigue Inventory for MS (WEIMuS), the Center for Epidemiologic Studies depression scale (CES-D), and the Paced Auditory Serial Addition Test (PASAT). Minimal clinically important difference (MCID) was evaluated for each test. Results A total of 189 participants were included: 122 were women (64.55%), with a mean age of 53.55 (±10.83). RFs showed significant improvement in all of the nonwalking outcomes (p < 0.05), except for a nonsignificant improvement in nondominant upper limb function and PASAT; the largest score improvement was seen in the physical and cognitive sections of the WEIMuS (25.69% and 29.81%, respectively, p < 0.001). Conclusion We provide evidence that physician's global judgement of walking improvement is a reliable measure for determining response to fampridine in nonwalking parameters, with fatigue showing the greatest score improvement after 2 weeks.
Collapse
Affiliation(s)
| | - Rocco Haase
- Center of Clinical Neuroscience, University Hospital, Carl Gustav Carus, TU Dresden, Germany
| | - Katja Akgün
- Center of Clinical Neuroscience, University Hospital, Carl Gustav Carus, TU Dresden, Germany
| | - Judith Eisele
- Center of Clinical Neuroscience, University Hospital, Carl Gustav Carus, TU Dresden, Germany
| | - Undine Proschmann
- Center of Clinical Neuroscience, University Hospital, Carl Gustav Carus, TU Dresden, Germany
| | - Thorsten Schultheiss
- Center of Clinical Neuroscience, University Hospital, Carl Gustav Carus, TU Dresden, Germany
| | - Raimar Kern
- Center of Clinical Neuroscience, University Hospital, Carl Gustav Carus, TU Dresden, Germany
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Fetscherstr. 74, Dresden, 01307, Germany
| |
Collapse
|
87
|
Farrell R. Clinical commentary on “De novo convulsive status epilepticus in patients with multiple sclerosis treated with dalfampridine”. Mult Scler 2019; 25:621-622. [DOI: 10.1177/1352458518810661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Rachel Farrell
- University College London Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
88
|
Kanhai KMS, Nij Bijvank JA, Wagenaar YL, Klaassen ES, Lim K, Bergheanu SC, Petzold A, Verma A, Hesterman J, Wattjes MP, Uitdehaag BMJ, van Rijn LJ, Groeneveld GJ. Treatment of internuclear ophthalmoparesis in multiple sclerosis with fampridine: A randomized double-blind, placebo-controlled cross-over trial. CNS Neurosci Ther 2019; 25:697-703. [PMID: 30756475 PMCID: PMC6515699 DOI: 10.1111/cns.13096] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/08/2018] [Indexed: 01/08/2023] Open
Abstract
Aim To examine whether the velocity of saccadic eye movements in internuclear ophthalmoparesis (INO) improves with fampridine treatment in patients with multiple sclerosis (MS). Methods Randomized, double‐blind, placebo‐controlled, cross‐over trial with fampridine in patients with MS and INO. Horizontal saccades were recorded at baseline and at multiple time points post‐dose. Main outcome measures were the change of peak velocity versional dysconjugacy index (PV‐VDI) and first‐pass amplitude VDI (FPA‐VDI). Both parameters were compared between fampridine and placebo using a mixed model analysis of variance taking patients as their own control. Pharmacokinetics was determined by serial blood sampling. Results Thirteen patients had a bilateral and 10 had a unilateral INO. One patient had an INO of abduction (posterior INO of Lutz) and was excluded. Fampridine significantly reduced both PV‐VDI (−17.4%, 95% CI: −22.4%, −12.1%; P < 0.0001) and FPA‐VDI (−12.5%, 95% CI: −18.9%, −5.5%; P < 0.01). Pharmacokinetics demonstrated that testing coincided with the average tmax at 2.08 hours (SD 45 minutes). The main adverse event reported after administration of fampridine was dizziness (61%). Conclusion Fampridine improves saccadic eye movements due to INO in MS. Treatment response to fampridine may gauge patient selection for inclusion to remyelination strategies in MS using saccadic eye movements as primary outcome measure.
Collapse
Affiliation(s)
| | - Jenny A Nij Bijvank
- Department of Ophthalmology, Neuro-ophthalmology Expertise Center, Amsterdam UMC, Amsterdam, the Netherlands.,Department of Neurology, MS Center and Neuro-ophthalmology Expertise Center, Amsterdam UMC, Amsterdam, the Netherlands
| | | | | | - KyoungSoo Lim
- Centre for Human Drug Research, Leiden, the Netherlands.,KCRN Research, Germantown, Maryland
| | | | - Axel Petzold
- Department of Ophthalmology, Neuro-ophthalmology Expertise Center, Amsterdam UMC, Amsterdam, the Netherlands.,Department of Neurology, MS Center and Neuro-ophthalmology Expertise Center, Amsterdam UMC, Amsterdam, the Netherlands.,The National Hospital for Neurology and Neurosurgery, Queen Square and Moorfields Eye Hospital, London, UK
| | - Ajay Verma
- Experimental Medicine, Biogen, Cambridge, Massachusetts
| | | | - Mike P Wattjes
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Bernard M J Uitdehaag
- Department of Neurology, MS Center and Neuro-ophthalmology Expertise Center, Amsterdam UMC, Amsterdam, the Netherlands
| | - Laurentius J van Rijn
- Department of Ophthalmology, Neuro-ophthalmology Expertise Center, Amsterdam UMC, Amsterdam, the Netherlands.,Department of Ophthalmology, Onze Lieve Vrouwe Gasthuis, Amsterdam, the Netherlands
| | - Geert Jan Groeneveld
- Centre for Human Drug Research, Leiden, the Netherlands.,Department of Neurology, MS Center and Neuro-ophthalmology Expertise Center, Amsterdam UMC, Amsterdam, the Netherlands
| |
Collapse
|
89
|
Decavel P, Moulin T, Sagawa Y. Gait tests in multiple sclerosis: Reliability and cut-off values. Gait Posture 2019; 67:37-42. [PMID: 30269001 DOI: 10.1016/j.gaitpost.2018.09.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 09/19/2018] [Accepted: 09/20/2018] [Indexed: 02/02/2023]
Abstract
BACKGROUND Gait limitation is one of the most common disabilities in people with multiple sclerosis (MS). Several studies have used gait parameters to determine the effects of different therapies. However, few studies have determined their reproducibility, also the therapeutic effects could be overestimated. RESEARCH QUESTION To examine the reproducibility in gait measurements during short and long distances. METHODS In this cross-sectional study we recruited a group of MS patients and compare it to a control group. The participants performed the following tests in a fixed order: a 25-foot walk at a comfortable speed, at a fast speed and during a dual task, a timed up-and-go test (TUG) and a six- minute walk test (6MWT). Two measurements were conducted a week apart. Systematic error was evaluated by the Student t-test, reliability by the intra-class correlation coefficients (ICC) and agreement by the minimum detectable change (MDC95). RESULTS A total of 58 people with MS and 19 healthy people were included. The absence of systematic error was only found for the fast speed condition. The reliability of the gait parameters had moderate to high ICC values (ICC > 0.7) except for the dual task cost (DTC) which was 0.45. The MDC95 was higher in people with MS compared to healthy people, and it was higher in people with MS for gait speeds in all conditions (> 34%). For the TUG and 6MWT, the MDC95 were 51.5% and 31.7% respectively. For people with MS the smallest MDC95 was found for the stance time for all conditions (6.8%), whereas the highest was found for the dual task cost (158.7%). SIGNIFICANCE The MDC95 values were higher than the cut-off point based on the minimally important clinical difference (MICD) proposed in previous studies. Thus, the MDC95 should be used as a cut-off rather than MICD values.
Collapse
Affiliation(s)
- Pierre Decavel
- Laboratoire d'Exploration Fonctionnelle Clinique du Mouvement, University Hospital of Besançon, F-25000, Besançon, France; Integrative and Clinical Neurosciences EA481, Bourgogne Franche-Comte University, F-25000, Besançon, France.
| | - Thierry Moulin
- Department of Neurology, University Hospital of Besançon, F-25000, Besançon, France; Integrative and Clinical Neurosciences EA481, Bourgogne Franche-Comte University, F-25000, Besançon, France
| | - Yoshimasa Sagawa
- Laboratoire d'Exploration Fonctionnelle Clinique du Mouvement, University Hospital of Besançon, F-25000, Besançon, France; Integrative and Clinical Neurosciences EA481, Bourgogne Franche-Comte University, F-25000, Besançon, France
| |
Collapse
|
90
|
Gross RH. The Multiple Sclerosis Severity Score: fluctuations and prognostic ability in a longitudinal cohort of patients with MS. Mult Scler J Exp Transl Clin 2019; 5:2055217319837254. [PMID: 30911402 PMCID: PMC6423683 DOI: 10.1177/2055217319837254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/19/2019] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The Multiple Sclerosis Severity Score (MSSS), combining the Expanded Disability Status Scale (EDSS) and disease duration, attempts to stratify multiple sclerosis (MS) patients based on their rate of progression. Its prognostic ability in the individual patient remains unproven. OBJECTIVES To assess the stability of MSSS within individual persons with MS in a longitudinal cohort, to evaluate whether certain factors influence MSSS variability, and to explore the ability of MSSS to predict future ambulatory function. METHODS A single-center retrospective review was performed of patients following a single provider for at least 8 years. Mixed model regression modeled MSSS over time. A Kaplan-Meier survival plot was fitted, using change of baseline MSSS by at least one decile as the event. Cox modeling assessed the influence of baseline clinical and demographic factors on the hazard of changing MSSS by at least one decile. Linear models evaluated the impact of baseline EDSS, baseline MSSS, and other factors on the Timed 25-Foot Walk (T25FW). RESULTS Out of 122 patients, 68 (55.7%) deviated from baseline MSSS by at least one decile. Final T25FW had slightly weaker correlation to baseline MSSS than to baseline EDSS, which was moderately strongly correlated with future log T25FW. CONCLUSION Individual MSSS scores often vary over time. Clinicians should exercise caution when using MSSS to prognosticate.
Collapse
Affiliation(s)
- RH Gross
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO,
USA
| |
Collapse
|
91
|
Hobart J, Ziemssen T, Feys P, Linnebank M, Goodman AD, Farrell R, Hupperts R, Blight AR, Englishby V, McNeill M, Chang I, Lima G, Elkins J, On behalf of the ENHANCE study investigators. Assessment of Clinically Meaningful Improvements in Self-Reported Walking Ability in Participants with Multiple Sclerosis: Results from the Randomized, Double-Blind, Phase III ENHANCE Trial of Prolonged-Release Fampridine. CNS Drugs 2019; 33:61-79. [PMID: 30535670 PMCID: PMC6328522 DOI: 10.1007/s40263-018-0586-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Walking impairment is a hallmark of multiple sclerosis (MS). It affects > 90% of individuals over time, reducing independence and negatively impacting health-related quality of life, productivity, and daily activities. Walking impairment is consistently reported as one of the most distressing impairments by individuals with MS. Prolonged-release (PR)-fampridine previously has been shown to improve objectively measured walking speed in walking-impaired adults with MS. The impact of PR-fampridine from the perspective of the individual with MS warrants full and detailed examination. OBJECTIVE The objective of this study was to evaluate whether PR-fampridine has a clinically meaningful effect on self-reported walking ability in walking-impaired participants with MS. METHODS ENHANCE was a phase III, randomized, double-blind, placebo-controlled study of PR-fampridine 10 mg twice daily in walking-impaired individuals age 18-70 years with either relapsing or progressive forms of MS and an Expanded Disability Status Scale (EDSS) score of 4.0-7.0 at screening. Participants were stratified by EDSS score (≤ 6.0 or 6.5-7.0) at randomization to ensure a balanced level of disability in the treatment groups. The primary endpoint was the proportion of participants with a mean improvement in the 12-item Multiple Sclerosis Walking Scale (MSWS-12) score exceeding the predefined threshold for clinically meaningful improvement (≥ 8 points) over 24 weeks. Secondary endpoints included the proportion with ≥ 15% improvement in Timed Up and Go (TUG) speed, and mean changes in Multiple Sclerosis Impact Scale physical impact subscale (MSIS-29 PHYS), Berg Balance Scale (BBS), and ABILHAND scores over 24 weeks. RESULTS In total, 636 participants with MS were randomized (PR-fampridine, n = 317; placebo, n = 319; modified intention-to-treat sample: PR-fampridine, n = 315; placebo, n = 318). At baseline in the PR-fampridine and placebo groups, 46% and 51% had a progressive form of MS, median [range] EDSS scores were 6.0 [4.0-7.0] and 5.5 [4.0-7.0], mean [range] MSWS-12 scores were 63.6 [0-100] and 65.4 [0-100], and mean [range] TUG speed was 0.38 [0.0-1.0] and 0.38 [0.0-1.2] feet/s, respectively. A significantly higher percentage of PR-fampridine-treated participants (136/315 [43.2%]) had clinically meaningful improvement in MSWS-12 score over 24 weeks versus placebo (107/318 [33.6%]; odds ratio 1.61 [95% confidence interval 1.15-2.26]; p = 0.006). For PR-fampridine versus placebo, significantly more participants had a ≥ 15% improvement in TUG speed, and there was significantly greater mean improvement in MSIS-29 PHYS score (p < 0.05); numerical improvements that were not statistically significant were observed in BBS/ABILHAND. Adverse events that were more common in the PR-fampridine group than placebo group (difference ≥ 3%) by Medical Dictionary for Regulatory Activities (MedDRA®) Preferred Term were urinary tract infection and insomnia. There were no seizures reported. CONCLUSIONS PR-fampridine treatment resulted in sustained, clinically meaningful improvements over 24 weeks in self-reported walking and functional ability in walking-disabled participants with MS. CLINICALTRIALS. GOV IDENTIFIER NCT02219932.
Collapse
Affiliation(s)
- Jeremy Hobart
- Plymouth University Peninsula Schools of Medicine and Dentistry, University Hospitals Plymouth NHS Trust, N13 ITTC Building, Plymouth Science Park, Plymouth, Devon, PL6 8BX, UK.
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Carl Gustav Carus University Clinic, Technical University of Dresden, Dresden, Germany
| | - Peter Feys
- BIOMED-REVAL, University of Hasselt, Diepenbeek, Belgium
| | - Michael Linnebank
- HELIOS Klinik Hagen-Ambrock, University Witten/Herdecke, Hagen, Germany
| | - Andrew D. Goodman
- School of Medicine and Dentistry, University of Rochester, Rochester, NY USA
| | - Rachel Farrell
- National Hospital for Neurology and Neurosurgery, University College London Hospitals and University College London Institute of Neurology, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Satchidanand N, Drake A, Smerbeck A, Hojnacki D, Kolb C, Patrick K, Weinstock-Guttman B, Motl R, Benedict RHB. Dalfampridine benefits ambulation but not cognition in multiple sclerosis. Mult Scler 2018; 26:91-98. [DOI: 10.1177/1352458518815795] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Background: Impaired cognition and ambulation are common in multiple sclerosis (MS). Dalfampridine is the first Food and Drug Administration (FDA)–approved medication to treat impaired ambulation in MS. Dalfampridine may benefit patients with cognitive impairment, given its effects on saltatory conduction and the association between cognitive and motor function. Objective: To examine the effects of dalfampridine on cognition in MS. To determine if the anticipated improved cognition is grounded in dalfampridine’s effects on ambulation. Methods: Adults with MS were randomized to dalfampridine ( n = 45) or placebo ( n = 16) for 12 weeks. Cognition and motor function were assessed at baseline and end-point. Results: T25FW and 6-minute walk (6MW) performance improved at end-point in the treatment group but not in the placebo group ( p < 0.05). Our primary outcome, performance on the Symbol Digit Modalities Test, did not improve. About 30% ( n = 12) of the dalfampridine group demonstrated ⩾20% improved ambulation and were categorized “responders.” Among “responders”, Symbol Digit Modalities test performance did not improve. However, performance on the Paced Auditory Serial Addition Test improved among “responders” ( p < 0.05). Conclusion: Dalfampridine benefits timed ambulation but not cognition. Some improvement among ambulation “responders” is consistent with prior reports of cognition-motor coupling in MS ( ClinicalTrials.gov #: NCT02006160).
Collapse
Affiliation(s)
- Nikhil Satchidanand
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Allison Drake
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - A Smerbeck
- Rochester Institute of Technology, Rochester, NY, USA
| | - David Hojnacki
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Channa Kolb
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Kara Patrick
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Bianca Weinstock-Guttman
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Robert Motl
- Department of Physical Therapy, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ralph HB Benedict
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| |
Collapse
|
93
|
Abstract
Multiple sclerosis (MS) is the most common chronic inflammatory, demyelinating and neurodegenerative disease of the central nervous system in young adults. This disorder is a heterogeneous, multifactorial, immune-mediated disease that is influenced by both genetic and environmental factors. In most patients, reversible episodes of neurological dysfunction lasting several days or weeks characterize the initial stages of the disease (that is, clinically isolated syndrome and relapsing-remitting MS). Over time, irreversible clinical and cognitive deficits develop. A minority of patients have a progressive disease course from the onset. The pathological hallmark of MS is the formation of demyelinating lesions in the brain and spinal cord, which can be associated with neuro-axonal damage. Focal lesions are thought to be caused by the infiltration of immune cells, including T cells, B cells and myeloid cells, into the central nervous system parenchyma, with associated injury. MS is associated with a substantial burden on society owing to the high cost of the available treatments and poorer employment prospects and job retention for patients and their caregivers.
Collapse
Affiliation(s)
- Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy. .,Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.
| | - Amit Bar-Or
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.,Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.,Neuroimmunology Unit, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Paolo Preziosa
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.,Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandra Solari
- Unit of Neuroepidemiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sandra Vukusic
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuro-inflammation, Fondation Eugène Devic EDMUS Contre la Sclérose en Plaques, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Lyon, France
| | - Maria A Rocca
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.,Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
94
|
Plummer P, Bohling CJ, Nickles LE, Williams AA, Thomas A, Markovic-Plese S, Giesser B. Case Report: Combining Dalfampridine with Multicomponent Exercise and Gait Training in a Person with Multiple Sclerosis. Int J MS Care 2018; 20:238-243. [PMID: 30374254 DOI: 10.7224/1537-2073.2017-074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background Dalfampridine extended release (D-ER) improves gait speed in some people with multiple sclerosis (MS), but many patients who take D-ER demonstrate only small improvements of questionable clinical significance. Physical therapy (PT) may augment the treatment effects of D-ER on the nervous system and improve clinical outcomes. This case report describes the successful use of D-ER combined with multicomponent PT in a patient who did not have a clinically important change in gait speed with D-ER alone. Methods A 59-year-old woman with a 6-year history of relapsing-remitting MS was prescribed D-ER by her neurologist. After 3 weeks of D-ER therapy (10 mg twice daily), she demonstrated only a 7.1% improvement in the Timed 25-Foot Walk test. She then commenced PT consisting of two 40-minute sessions per week for 6 weeks while continuing D-ER therapy. Training focused on gait, balance, coordination, functional strengthening, and dual-task performance. Results After 6 weeks of D-ER + PT, she had a further 14.6% improvement in Timed 25-Foot Walk gait speed, for a total improvement of 20.7%, which elevated her above the clinically meaningful threshold of 20%. Similar patterns of improvement were also observed for self-selected gait speed in single- and dual-task conditions. Improvements in fast and dual-task gait speed were retained 3 weeks later. Conclusions For this patient, combining PT with D-ER therapy improved gait speed more than the use of D-ER alone. Further investigation of D-ER + PT or PT as an alternative to D-ER in patients with submeaningful medication response is warranted.
Collapse
|
95
|
Panicucci E, Cohen M, Bourg V, Rocher F, Thomas P, Lebrun C. De novo convulsive status epilepticus in patients with multiple sclerosis treated with dalfampridine. Mult Scler 2018; 25:618-621. [PMID: 30375922 DOI: 10.1177/1352458518790379] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Dalfampridine extended release (DAL) is a broad-spectrum voltage-gated potassium channel blocker that is indicated in multiple sclerosis to improve the nerve conduction of demyelinated axons. Seizures are a known side effect of DAL, which is contraindicated in patients with a history of epilepsy. OBJECTIVE Three cases of multiple sclerosis (MS) with de novo convulsive status epilepticus (CSE) probably related to dalfampridine administration are described. METHODS No patients had a history of seizures or renal impairment. Biological tests were normal. A brain magnetic resonance imaging (MRI) showed diffuse cortical and subcortical atrophy without active inflammatory lesions. RESULTS All three patients presented with CSE that was attributed to DAL and so was discontinued. CONCLUSION These case reports illustrate that, aside from seizures, de novo CSE is a potential complication of MS patients treated with DAL.
Collapse
Affiliation(s)
| | - Mikael Cohen
- Service de Neurologie, CRCSEP, Hôpital Pasteur 2, Nice, France
| | - Veronique Bourg
- Service de Neurologie, CRCSEP, Hôpital Pasteur 2, Nice, France
| | - Fanny Rocher
- Centre régional de Pharmacovigilance, Hôpital Cimiez, Nice, France
| | - Pierre Thomas
- Service de Neurologie, CRCSEP, Hôpital Pasteur 2, Nice, France
| | | |
Collapse
|
96
|
Effect of patients' expectations on clinical response to fampridine treatment. Neurol Sci 2018; 40:175-180. [PMID: 30374581 DOI: 10.1007/s10072-018-3613-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 10/18/2018] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Patient expectation of treatment outcome is one of the primary mechanisms underlying the placebo effect. In multiple sclerosis trials with symptomatic treatments, a robust placebo effect is observed, which might be related to patient expectations. The aim of this study was to evaluate whether patient expectations regarding fampridine treatment influence the clinical response after 4 weeks and 6 months of treatment. MATERIALS AND METHODS We designed and carried out a prospective study from June 2015 to August 2017. Before treatment, patients completed a questionnaire including a scale evaluating their expectations regarding the treatment. The effect of baseline positive expectancy on the response status after 4 weeks and 6 months of treatment was analyzed through univariable and, when applicable, multivariable analysis. RESULTS A total of 47 consecutive patients were included in the study. At week 4, 37 (78.7%) patients were classified as responders; a one-point increase in the positive expectancy questionnaire was significantly associated with a fourfold increase in the likelihood of being a responder [OR = 4.020 (95% CI 1.082-14.933); p = 0.038]. At 6 months, 43 patients completed follow-up. The number of responders decreased to 28; at this point, positive expectancy at baseline was no longer associated with response status. CONCLUSION Baseline positive expectancy regarding fampridine was determinant of the clinical response after 4 weeks of treatment. However, in the long term, fampridine efficacy was not dependent on expectations prior to treatment.
Collapse
|
97
|
Abstract
Multiple sclerosis (MS) is a chronic neurologic disease and the most common cause of nontraumatic disability in young adults in the United States. It strikes adults in the prime of life and can gradually diminish physical and mental function, affect relationships, employment, financial capacity, and independence. Neurologic symptoms fluctuate in a pattern of remissions and exacerbations for the majority of those with MS. A multidisciplinary treatment approach is essential. There are a wide number of medications that can be used. Treating the disease at an early stage within the first 5 years of clinical symptom onset is optimal. Home healthcare clinicians can use assessment tools to evaluate disease progression, teach the patient and family how to live with the disorder, and monitor the patient for medication side effects.
Collapse
|
98
|
Filli L, Werner J, Beyer G, Reuter K, Petersen JA, Weller M, Zörner B, Linnebank M. Predicting responsiveness to fampridine in gait-impaired patients with multiple sclerosis. Eur J Neurol 2018; 26:281-289. [DOI: 10.1111/ene.13805] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/28/2018] [Indexed: 11/29/2022]
Affiliation(s)
- L. Filli
- Department of Neurology; University Hospital Zurich; Zurich
| | - J. Werner
- Department of Neurology; University Hospital Zurich; Zurich
| | - G. Beyer
- Department of Neurology; University Hospital Zurich; Zurich
| | - K. Reuter
- Department of Neurology; University Hospital Zurich; Zurich
| | - J. A. Petersen
- Department of Neurology; University Hospital Zurich; Zurich
| | - M. Weller
- Department of Neurology; University Hospital Zurich; Zurich
| | - B. Zörner
- Spinal Cord Injury Center; Balgrist University Hospital; Zurich Switzerland
| | - M. Linnebank
- Department of Neurology; University Hospital Zurich; Zurich
- Department of Neurology; Helios-Klinik Hagen-Ambrock; University Witten/Herdecke; Hagen Germany
| |
Collapse
|
99
|
Albrecht P, Bjørnå IK, Brassat D, Farrell R, Feys P, Hobart J, Hupperts R, Linnebank M, Magdič J, Oreja-Guevara C, Pozzilli C, Salgado AV, Ziemssen T. Prolonged-release fampridine in multiple sclerosis: clinical data and real-world experience. Report of an expert meeting. Ther Adv Neurol Disord 2018; 11:1756286418803248. [PMID: 30305850 PMCID: PMC6174649 DOI: 10.1177/1756286418803248] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/25/2018] [Indexed: 01/07/2023] Open
Abstract
Prolonged-release (PR) fampridine is the only approved medication to improve walking in multiple sclerosis (MS), having been shown to produce a clinically meaningful improvement in walking ability in the subset of MS patients with Expanded Disability Status Scale 4-7. Recent responder subgroup analyses in the phase III ENHANCE study show a large effect size in terms of an increase of 20.58 points on the patient-reported 12-item MS Walking Scale in the 43% of patients classified as responders to PR-fampridine, corresponding to a standardized response mean of 1.68. Use of PR-fampridine in clinical practice varies across Europe, depending partly on whether it is reimbursed. A group of European MS experts met in June 2017 to discuss their experience with using PR-fampridine, including their views on the patient population for treatment, assessment of treatment response, re-testing and re-treatment, and stopping criteria. This article summarizes the experts' opinions on how PR-fampridine can be used in real-world clinical practice to optimize the benefits to people with MS with impaired walking ability.
Collapse
Affiliation(s)
- Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich Heine University, Moorenstr. 5, 40225 Duesseldorf, Germany
| | | | - David Brassat
- Centre de Resource et de Competence-SEP CHU-Toulouse et UMR1043, Université de Toulouse III, Toulouse, France
| | - Rachel Farrell
- Department of Neuroinflammation, UCL Institute of Neurology, London and National Hospital for Neurology and Neurosurgery, UCLH NHS Foundation Trust, London, UK
| | - Peter Feys
- REVAL/BIOMED, Faculty of Rehabilitation Sciences, Hasselt University, Hasselt, Belgium
| | - Jeremy Hobart
- Department of Clinical Trials and Health Research: Translational & Stratified Medicine, Plymouth
| | - Raymond Hupperts
- Department of Neurology, Zuyderland Medical Center, Sittard-Geleen, Netherlands
| | - Michael Linnebank
- Department of Neurology, Helios Clinic Hagen-Ambrock, Hagen, Germany
| | - Jožef Magdič
- Department of Neurology, University Medical Center Maribor, Maribor, Slovenia
| | - Celia Oreja-Guevara
- Neurología, Hospital Clínico San Carlos, Departamento de Medicina, Universidad Complutense de Madrid, Spain
| | - Carlo Pozzilli
- Department of Neurology, University La Sapienza, Rome, Italy
| | | | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, MS Center Dresden, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| |
Collapse
|
100
|
Bakirtzis C, Konstantinopoulou E, Langdon DW, Grigoriadou E, Minti F, Nikolaidis I, Boziki MK, Tatsi T, Ioannidis P, Karapanayiotides T, Afrantou T, Hadjigeorgiou G, Grigoriadis N. Long-term effects of prolonged-release fampridine in cognitive function, fatigue, mood and quality of life of MS patients: The IGNITE study. J Neurol Sci 2018; 395:106-112. [PMID: 30308444 DOI: 10.1016/j.jns.2018.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/13/2018] [Accepted: 10/02/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Studies have reported conflicting results regarding the potential benefit of prolonged release (PR) fampridine in other domains besides walking. Moreover, only a small number of studies have explored long- term effects of PR fampridine. The aim of this study was to assess cognitive function, quality of life, mood and fatigue in MS patients treated with fampridine after 6 and 12 months of treatment. METHODS IGNITE was an observational, open label study. Subjects were examined with the timed 25-ft walk (T25FW) and the BICAMS battery and were asked to complete the Multiple Sclerosis Impact Scale (MSIS-29), Modified Fatigue Impact Scale (MFIS), Beck Depression Inventory-II (BDI-II) and MS International Quality-of-Life questionnaire (MUSIQOL) at baseline and at weeks 24 and 48. Patients were sub-grouped into responders (n:40) and non-responders (n:20) according to T25FW performance after 2 weeks on treatment. RESULTS After 6 months, statistically significant improvement was observed on T25FW (p < .001), SDMT (p < .001) and MSIS29 (p < .001), for responders. After 1 year on treatment, statistically significant improvement was observed in T25FW (p < .001), MSIS29 (p = .004), SDMT (p < .001) and MUSIQOL (p = .03) for responders. There were no statistically significant improvements for the non-responders. CONCLUSIONS PR Fampridine may have a beneficial effect on information processing speed though not on memory. Study data provide some evidence that fampridine treatment may reduce the impact of MS on daily activities and improve quality of life but has no effect on subjective fatigue and mood.
Collapse
Affiliation(s)
- C Bakirtzis
- The Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece.
| | - E Konstantinopoulou
- Lab of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - D W Langdon
- Royal Holloway, University of London, London, UK
| | - E Grigoriadou
- The Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - F Minti
- The Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - I Nikolaidis
- The Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - M K Boziki
- The Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - T Tatsi
- The Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - P Ioannidis
- The Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - T Karapanayiotides
- The Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - T Afrantou
- The Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - G Hadjigeorgiou
- Department of Neurology, Medical School, University of Cyprus, Cyprus
| | - N Grigoriadis
- The Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| |
Collapse
|