Michael C Levin, Sangmin Lee, Lidia A Gardner, Yoojin Shin, Joshua N Douglas, Chassidy J Groover
{"title":"Pathogenic mechanisms of neurodegeneration based on the phenotypic expression of progressive forms of immune-mediated neurologic disease.","authors":"Michael C Levin, Sangmin Lee, Lidia A Gardner, Yoojin Shin, Joshua N Douglas, Chassidy J Groover","doi":"10.2147/DNND.S38353","DOIUrl":"10.2147/DNND.S38353","url":null,"abstract":"<p><p>Considering there are no treatments for progressive forms of multiple sclerosis (MS), a comprehensive understanding of the role of neurodegeneration in the pathogenesis of MS should lead to novel therapeutic strategies to treat it. Many studies have implicated viral triggers as a cause of MS, yet no single virus has been exclusively shown to cause MS. Given this, human and animal viral models of MS are used to study its pathogenesis. One example is human T-lymphotropic virus type 1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Importantly, HAM/TSP is similar clinically, pathologically, and immunologically to progressive MS. Interestingly, both MS and HAM/TSP patients were found to make antibodies to heterogeneous nuclear ribonucleoprotein (hnRNP) A1, an RNA-binding protein overexpressed in neurons. Anti-hnRNP A1 antibodies reduced neuronal firing and caused neurodegeneration in neuronal cell lines, suggesting the autoantibodies are pathogenic. Further, microarray analyses of neurons exposed to anti-hnRNP A1 antibodies revealed novel pathways of neurodegeneration related to alterations of RNA levels of the spinal paraplegia genes (SPGs). Mutations in SPGs cause hereditary spastic paraparesis, genetic disorders clinically indistinguishable from progressive MS and HAM/TSP. Thus, there is a strong association between involvement of SPGs in neurodegeneration and the clinical phenotype of progressive MS and HAM/TSP patients, who commonly develop spastic paraparesis. Taken together, these data begin to clarify mechanisms of neurodegeneration related to the clinical presentation of patients with chronic immune-mediated neurological disease of the central nervous system, which will give insights into the design of novel therapies to treat these neurological diseases.</p>","PeriodicalId":11147,"journal":{"name":"Degenerative Neurological and Neuromuscular Disease","volume":"2 ","pages":"175-187"},"PeriodicalIF":0.0,"publicationDate":"2012-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6065584/pdf/dnnd-2-175.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37073184","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Evaluation of tafamidis as first-line therapeutic agent for transthyretin familial amyloidotic polyneuropathy.","authors":"Joel N Buxbaum","doi":"10.2147/DNND.S24624","DOIUrl":"https://doi.org/10.2147/DNND.S24624","url":null,"abstract":"<p><p>Almost 100 mutations in the human transthyretin (TTR) gene cause the autosomal dominant disorders of familial amyloidotic polyneuropathy (FAP) and familial amyloidotic cardiomyopathy. While these have been clinically classified as separate disorders, the peripheral and autonomic nervous systems and the heart are frequently involved in the same patient. Deposition of amyloid derived from a kinetically or thermodynamically unstable mutant TTR precursor produces an ascending sensorimotor polyneuropathy with marked autonomic involvement. Since 1990, treatment has been liver transplantation from a donor carrying two wild-type TTR genes, providing a crude form of gene therapy. Multiple studies have shown that small molecules fitting in the T<sub>4</sub>-binding pocket of TTR can stabilize the molecule, reducing its capacity to release the fibril precursor. Tafamidis is the first molecule to be tested in a placebo-controlled trial in patients with TTR-associated FAP. While the trial did not achieve its primary endpoints, it did stabilize TTR in vivo and had a favorable effect on some aspects of disease progression, particularly when administered early in the course. It may represent an alternative to liver transplantation, particularly in patients with early disease related to the V30M mutation. Longer-term studies are required to determine whether it represents a stabilizing or remittive form of treatment.</p>","PeriodicalId":11147,"journal":{"name":"Degenerative Neurological and Neuromuscular Disease","volume":"2 ","pages":"165-173"},"PeriodicalIF":0.0,"publicationDate":"2012-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/DNND.S24624","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37073181","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Recent advances in Duchenne muscular dystrophy.","authors":"Kelly J Perkins, Kay E Davies","doi":"10.2147/DNND.S26637","DOIUrl":"https://doi.org/10.2147/DNND.S26637","url":null,"abstract":"<p><p>Duchenne muscular dystrophy (DMD), an allelic X-linked progressive muscle-wasting disease, is one of the most common single-gene disorders in the developed world. Despite knowledge of the underlying genetic causation and resultant pathophysiology from lack of dystrophin protein at the muscle sarcolemma, clinical intervention is currently restricted to symptom management. In recent years, however, unprecedented advances in strategies devised to correct the primary defect through gene- and cell-based therapeutics hold particular promise for treating dystrophic muscle. Conventional gene replacement and endogenous modification strategies have greatly benefited from continued improvements in encapsidation capacity, transduction efficiency, and systemic delivery. In particular, RNA-based modifying approaches such as exon skipping enable expression of a shorter but functional dystrophin protein and rapid progress toward clinical application. Emerging combined gene- and cell-therapy strategies also illustrate particular promise in enabling ex vivo genetic correction and autologous transplantation to circumvent a number of immune challenges. These approaches are complemented by a vast array of pharmacological approaches, in particular the successful identification of molecules that enable functional replacement or ameliorate secondary DMD pathology. Animal models have been instrumental in providing proof of principle for many of these strategies, leading to several recent trials that have investigated their efficacy in DMD patients. Although none has reached the point of clinical use, rapid improvements in experimental technology and design draw this goal ever closer. Here, we review therapeutic approaches to DMD, with particular emphasis on recent progress in strategic development, preclinical evaluation and establishment of clinical efficacy. Further, we discuss the numerous challenges faced and synergistic approaches being devised to combat dystrophic pathology effectively.</p>","PeriodicalId":11147,"journal":{"name":"Degenerative Neurological and Neuromuscular Disease","volume":"2 ","pages":"141-164"},"PeriodicalIF":0.0,"publicationDate":"2012-10-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/DNND.S26637","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37073182","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Optimal management for people with severe spasticity.","authors":"Jeffrey S Shilt, Pennie S Seibert, Vivek Kadyan","doi":"10.2147/DNND.S16630","DOIUrl":"https://doi.org/10.2147/DNND.S16630","url":null,"abstract":"<p><p>Spasticity is characterized by velocity-dependent increase in tonic stretch reflexes and tendon jerks. Many people affected by spasticity receive late treatment, or no treatment, which greatly reduces the potential to regain full motor control and restore function. There is much to consider before determining treatment for people with spasticity. Treatment of pediatric patients increases the complexity, because of the substantial difference between adult and pediatric spasticity. Proper patient evaluation, utilization of scales and measures, and obtaining patient and caregiver history is vital in determining optimal spasticity treatment. Further, taking into consideration the limitations and desires of individuals serve as a guide to best management. We have grouped contributing factors into the IDAHO Criteria to elucidate a multidisciplinary approach, which considers a person's complete field of experience. This model is applied to goal setting, and recognizes the importance of a spasticity management team, comprising the treatment subject, his/her family, the environment, and a supportive, well-informed medical staff. The criteria take into account the complexity associated with diagnosing and treating spasticity, with the ultimate goal of improved function.</p>","PeriodicalId":11147,"journal":{"name":"Degenerative Neurological and Neuromuscular Disease","volume":"2 ","pages":"133-140"},"PeriodicalIF":0.0,"publicationDate":"2012-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/DNND.S16630","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37073180","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Paolo Giannetti, Flavia Niccolini, Richard Nicholas
{"title":"BG-12 and its potential for the prevention of relapse in multiple sclerosis.","authors":"Paolo Giannetti, Flavia Niccolini, Richard Nicholas","doi":"10.2147/DNND.S35790","DOIUrl":"https://doi.org/10.2147/DNND.S35790","url":null,"abstract":"<p><p>Multiple sclerosis (MS) arises from an immune attack on the central nervous system producing demyelination and axonal loss. Clinically the relapsing-remitting course is characterized by subacute onset of neurological symptoms usually with partial or complete recovery, while the progressive course, predominant in the later stages, is characterized by progressive disability in the absence of relapses. A number of disease-modifying treatments have been developed and are increasingly effective at targeting relapses. Early injectable therapies such as interferon and glatiramer acetate are only partially effective, but have a good safety record. Recently, natalizumab, an intravenous therapy, demonstrated increased effectiveness, but side effects complicate its use. The first oral therapy offering good efficacy and convenience, fingolimod, was approved in USA in 2010 and Europe in 2011. BG-12 is a potential novel oral therapy for MS, which has previously been used as a different formulation for psoriasis. It has anti-inflammatory and neuroprotective actions in vitro, which makes it a promising candidate for future therapies. Phase II studies showed that BG-12 reduced MRI inflammatory activity over placebo, which was confirmed in two Phase III studies indicating immune modulation may be its principal action rather than neuroprotection. In these studies, BG-12 reduced relapse rates consistently with variable effects on progression and few serious adverse events. With its favorable efficacy-tolerability profile, BG-12 could offer a substantial step forward for the care for subjects affected by relapsing MS.</p>","PeriodicalId":11147,"journal":{"name":"Degenerative Neurological and Neuromuscular Disease","volume":"2 ","pages":"119-132"},"PeriodicalIF":0.0,"publicationDate":"2012-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/DNND.S35790","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37073179","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Nader Pouratian, Sandeep Thakkar, Won Kim, Jeff M Bronstein
{"title":"Deep brain stimulation for the treatment of Parkinson's disease: efficacy and safety.","authors":"Nader Pouratian, Sandeep Thakkar, Won Kim, Jeff M Bronstein","doi":"10.2147/DNND.S25750","DOIUrl":"https://doi.org/10.2147/DNND.S25750","url":null,"abstract":"<p><p>Deep brain stimulation (DBS) surgery has become increasingly utilized in the treatment of advanced Parkinson's disease. Over the past decade, a number of studies have demonstrated that DBS is superior to best medical management in appropriately selected patients. The primary targets for DBS in Parkinson's disease include the subthalamic nucleus and the internal segment of the globus pallidus, both of which improve the cardinal motor features in Parkinson's disease. Recent randomized studies have revealed that both targets are similarly effective in treating the motor symptoms of Parkinson's disease, but emerging evidence suggests that the globus pallidus may be the preferred target in many patients, based on differences in nonmotor outcomes. Here, we review appropriate patient selection, and the efficacy and safety of DBS therapy in Parkinson's disease. Best outcomes are achieved if the problems of the individual patient are considered when evaluating surgical candidates and considering whether the subthalamic nucleus or the globus pallidus internus should be targeted.</p>","PeriodicalId":11147,"journal":{"name":"Degenerative Neurological and Neuromuscular Disease","volume":"2012 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2012-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/DNND.S25750","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"31920812","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ole B Suhr, Sandra Gustavsson, Victoria Heldestad, Rolf Hörnsten, Per Lindqvist, Erik Nordh, Urban Wiklund
{"title":"New insights into the clinical evaluation of hereditary transthyretin amyloidosis patients: a single center's experience.","authors":"Ole B Suhr, Sandra Gustavsson, Victoria Heldestad, Rolf Hörnsten, Per Lindqvist, Erik Nordh, Urban Wiklund","doi":"10.2147/DNND.S24652","DOIUrl":"10.2147/DNND.S24652","url":null,"abstract":"<p><p>Over the last decade, new medical treatment modalities have emerged based on increased insights into amyloid formation. With the increased possibilities for treatment of amyloidosis caused by transthyretin (TTR) amyloid deposits comes the need for diagnostic procedures for early diagnosis and better tools to follow disease progression. This is of particular importance in clinical trials evaluating the efficacy of new treatments. Until recently, the treatment of TTR amyloidosis (ATTR) was based solely on liver transplantation, a procedure that has halted disease progression in many patients. Liver transplantation has been especially effective in patients under the age of 50 years carrying the TTR V30M mutation, whereas the outcome of the procedure has been variable for others, particularly elderly male patients and those carrying a non-V30M mutation. This review concentrates on new insights derived from our center's experience with liver transplantation, how to implement this experience in evaluation of new treatment modalities for ATTR, and how to facilitate early diagnosis of neuropathy with easily available diagnostic tools. Attention has focused on manifestations of the disease that involve the heart and the peripheral nervous system; change in peripheral nerve function has been the primary endpoint in two controlled clinical trials, one finished and one ongoing. New insights into the amyloid formation process and the lessons learned from liver transplantation give the opportunity to design potentially effective treatment modalities for ATTR. It appears reasonable to suspect that a combination of different treatment modalities may be required to treat the disease, and that different treatment regimes will be designed according to the phenotype of the disease. For the patients and their relatives there is now a solid foundation for optimism, with prospects of several effective medical treatment possibilities within the coming decade.</p>","PeriodicalId":11147,"journal":{"name":"Degenerative Neurological and Neuromuscular Disease","volume":"2 ","pages":"93-106"},"PeriodicalIF":0.0,"publicationDate":"2012-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/DNND.S24652","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37073178","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Stem cells and regenerative therapies for Parkinson's disease.","authors":"Krista Farrell, Roger A Barker","doi":"10.2147/DNND.S16087","DOIUrl":"https://doi.org/10.2147/DNND.S16087","url":null,"abstract":"<p><p>Currently the mainstay of Parkinson's disease (PD) therapy is the pharmacological replacement of the loss of the dopaminergic nigrostriatal pathway using drugs such as dopamine agonists and levodopa. Whilst these drugs effectively ameliorate some of the motor features of PD, they do not improve many of the nonmotor features that arise secondary to pathology outside of this system, nor do they slow the progressive neurodegeneration that is a characteristic of the disease. Regenerative therapies for PD seek to fill this therapeutic gap, with cell transplantation being the most explored approach to date. A number of different cell sources have been used in this therapeutic approach, but to date, the most successful has been the use of fetal ventral mesencephalic (VM) tissue that contains within it the developing nigral dopaminergic cells. Cell transplantation for PD was pioneered in the 1980-1990s, with several successful open-label trials of fetal VM transplantation in patients with relatively advanced PD. Whilst these findings were not replicated in two subsequent double-blind sham-surgery controlled trials, there were reasons to explain this outside of the one drawn at the time that these therapies are ineffective. Indeed all these studies have provided evidence that following the transplantation of fetal VM tissue, dopaminergic cells can survive long term, produce dopamine, and bring about clinical improvements in younger patients over many years. The use of fetal tissue, irrespective of its true efficacy, will never become a widely available therapy for PD for a host of practical and ethical reasons, and thus much work has been put in recently to exploring the utility of stem cells as a source of nigral dopaminergic neurons. In this respect, the advent of embryonic stem cell and induced pluripotent cells has heralded a new era in cell therapy for PD, and several groups have now demonstrated that these cells can form dopaminergic neurons which improve functional deficits in animal models of PD. Whilst encouraging, problems with respect to the immunogenicity and tumorigenicity of these cells means that they will need to be used in the clinic cautiously. Other regenerative therapies in PD have been tried over the years and include the use of trophic factors. This has primarily involved glial cell line-derived neurotrophic factor (GDNF) and again has produced mixed clinical effects, and in order to try and resolve this, a new trial of intraputamenal GDNF is now being planned. In addition, a new trial for platelet derived growth factor as a treatment for PD has just completed recruitment, and PYM50028 (Cogane) an oral agent shown in animal models to reduce the effects of MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) lesioning by the induction of growth factors is currently under investigation in a multicentre Phase II trial. Overall, there are a number of promising new regenerative therapies being developed and tested in PD, although the","PeriodicalId":11147,"journal":{"name":"Degenerative Neurological and Neuromuscular Disease","volume":"2 ","pages":"79-92"},"PeriodicalIF":0.0,"publicationDate":"2012-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/DNND.S16087","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37073177","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Cell adhesion molecules in Alzheimer's disease.","authors":"Malin Wennström, Henrietta M Nielsen","doi":"10.2147/DNND.S19829","DOIUrl":"https://doi.org/10.2147/DNND.S19829","url":null,"abstract":"<p><p>Cell adhesion molecules (CAMs) mediate interactions between cells and their surroundings that are vital to processes controlling for cell survival, activation, migration, and plasticity. However, increasing evidence suggests that CAMs also mediate mechanisms involved in several neurological diseases. This article reviews the current knowledge on the role of CAMs in amyloid-β (Aβ) metabolism, cell plasticity, neuroinflammation, and vascular changes, all of which are considered central to the pathogenesis and progression of Alzheimer's disease (AD). This paper also outlines the possible roles of CAMs in current and novel AD treatment strategies.</p>","PeriodicalId":11147,"journal":{"name":"Degenerative Neurological and Neuromuscular Disease","volume":"2 ","pages":"65-77"},"PeriodicalIF":0.0,"publicationDate":"2012-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/DNND.S19829","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37072203","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Walking impairment in patients with multiple sclerosis - a new therapeutic approach and clinical potential of dalfampridine extended release tablets.","authors":"Herbert R Henney, Andrew R Blight","doi":"10.2147/DNND.S19839","DOIUrl":"https://doi.org/10.2147/DNND.S19839","url":null,"abstract":"<p><p>Walking impairment is a clinical hallmark of multiple sclerosis (MS) that has been under-recognized as a therapeutic target for pharmacologic intervention. The development and approval of dalfampridine extended release tablets (dalfampridine-ER; known as prolonged-, modified, or sustained-release fampridine outside the USA), 10 mg taken twice daily, to improve walking in patients with MS, fills a previously unmet need. In three randomized, double-blind, placebo-controlled trials, dalfampridine-ER improved walking speed in approximately one-third (37%) of treated patients, and average walking speed on therapy among these responders improved by approximately 25% relative to baseline. Walking-speed improvement among responders was clinically significant, as determined by a statistically significant improvement in the patient-reported 12-item Multiple Sclerosis Walking Scale. Long-term extension studies indicate that responders were able to maintain benefits, compared with nonresponders over prolonged periods of treatment. Dalfampridine-ER was generally well tolerated. Dizziness, insomnia, balance disorder, headache, nausea, urinary tract infection, and asthenia were the most common adverse events. Although the incidence of seizures appeared to be dose related, among patients treated with dalfampridine-ER in the three trials, the rate of seizures was 0.25%. These efficacy and safety data suggest that dalfampridine-ER can be a useful and clinically relevant addition to the pharmacologic armamentarium for the management of MS symptoms and disabilities. Because of its narrow therapeutic index and potential for seizures, it is especially important in the clinical setting to adhere to the dosing recommended in the approved labels.</p>","PeriodicalId":11147,"journal":{"name":"Degenerative Neurological and Neuromuscular Disease","volume":"2 ","pages":"53-64"},"PeriodicalIF":0.0,"publicationDate":"2012-06-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/DNND.S19839","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37072200","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}