Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 208
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Dysphagia ; 37(3): 473-487, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34226958

RESUMEN

Autoimmune neurogenic dysphagia refers to manifestation of dysphagia due to autoimmune diseases affecting muscle, neuromuscular junction, nerves, roots, brainstem, or cortex. Dysphagia is either part of the evolving clinical symptomatology of an underlying neurological autoimmunity or occurs as a sole manifestation, acutely or insidiously. This opinion article reviews the autoimmune neurological causes of dysphagia, highlights clinical clues and laboratory testing that facilitate early diagnosis, especially when dysphagia is the presenting symptom, and outlines the most effective immunotherapeutic approaches. Dysphagia is common in inflammatory myopathies, most prominently in inclusion body myositis, and is frequent in myasthenia gravis, occurring early in bulbar-onset disease or during the course of progressive, generalized disease. Acute-onset dysphagia is often seen in Guillain-Barre syndrome variants and slowly progressive dysphagia in paraneoplastic neuropathies highlighted by the presence of specific autoantibodies. The most common causes of CNS autoimmune dysphagia are demyelinating and inflammatory lesions in the brainstem, occurring in patients with multiple sclerosis and neuromyelitis optica spectrum disorders. Less common, but often overlooked, is dysphagia in stiff-person syndrome especially in conjunction with cerebellar ataxia and high anti-GAD autoantibodies, and in gastrointestinal dysmotility syndromes associated with autoantibodies against the ganglionic acetyl-choline receptor. In the setting of many neurological autoimmunities, acute-onset or progressive dysphagia is a potentially treatable condition, requiring increased awareness for prompt diagnosis and early immunotherapy initiation.


Asunto(s)
Trastornos de Deglución , Síndrome de Guillain-Barré , Esclerosis Múltiple , Autoanticuerpos , Trastornos de Deglución/etiología , Humanos
2.
Curr Opin Neurol ; 33(5): 545-552, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32833750

RESUMEN

PURPOSE OF REVIEW: To provide an update on immunomodulating and immunosuppressive therapies in myasthenia gravis and highlight newly approved, or pending approval, therapies with new biologics. RECENT FINDINGS: Preoperative IVIg is not needed to prevent myasthenic crisis in stable myasthenia gravis patients scheduled for surgery under general anesthesia, based on controlled data. Rituximab, if initiated early in new-onset myasthenia gravis, can lead to faster and more sustained remission even without immunotherapies in 35% of patients at 2 years. Biomarkers determining the timing for follow-up infusions in Rituximab-responding AChR-positive patients are discussed. Most patients with MuSK-positive myasthenia gravis treated with Rituximab have sustained long-term remission with persistent reduction of IgG4 anti-MuSK antibodies. Eculizumb in the extension REGAIN study showed sustained long-term pharmacological remissions and reduced exacerbations. Three new biologic agents showed promising results in phase-II controlled myasthenia gravis trials: Zilucoplan, a subcutaneous macrocyclic peptide inhibiting complement C5; Efgartigimod, an IgG1-derived Fc fragment binding to neonatal FcRn receptor; and Rozanolixizumab, a high-affinity anti-FcRn monoclonal antibody. Finally, the safety of ongoing myasthenia gravis immunotherapies during COVID19 pandemic is discussed. SUMMARY: New biologics against B cells, complement and FcRn receptor, are bringing us closer to successful targeted immunotherapies in the chronic management of myasthenia gravis promising an exciting future for antibody-mediated neurological diseases.


Asunto(s)
Factores Inmunológicos/uso terapéutico , Inmunoterapia/métodos , Miastenia Gravis/tratamiento farmacológico , Autoanticuerpos/inmunología , COVID-19 , Humanos , Factores Inmunológicos/efectos adversos , Inmunoterapia/efectos adversos , Miastenia Gravis/inmunología , SARS-CoV-2
3.
J Autoimmun ; 104: 102339, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31611142

RESUMEN

Autoimmune encephalitides, with an estimated incidence of 1.5 per million population per year, although described only 15 years ago, have already had a remarkable impact in neurology and paved the field to autoimmune neuropsychiatry. Many patients traditionally presented with aberrant behavior, especially of acute or subacute onset, and treated with anti-psychotic therapies, turn out to have a CNS autoimmune disease with pathogenic autoantibodies against synaptic antigens responding to immunotherapies. The review describes the clinical spectrum of these disorders, and the pathogenetic role of key autoantibodies directed against: a) cell surface synaptic antigens and receptors, including NMDAR, GABAa, GABAb, AMPA and glycine receptors; b) channels such as AQP4 water-permeable channel or voltage-gated potassium channels; c) proteins that stabilize voltage-gated potassium channel complex into the membrane, like the LGI1 and CASPR2; and d) enzymes that catalyze the formation of neurotransmitters such as Glutamic Acid Decarboxylase (GAD). These antibodies, effectively target excitatory or inhibitory synapses in the limbic system, basal ganglia or brainstem altering synaptic function and resulting in uncontrolled neurological excitability disorder clinically manifested with psychosis, agitation, behavioral alterations, depression, sleep disturbances, seizure-like phenomena, movement disorders such as ataxia, chorea and dystonia, memory changes or coma. Some of the identified triggering factors include: viruses, especially herpes simplex, accounting for the majority of relapses occurring after viral encephalitis, which respond to immunotherapy rather than antiviral agents; tumors especially teratoma, SCLC and thymomas; and biological cancer therapies (immune-check-point inhibitors). As anti-synaptic antibodies persist after viral infections or tumor removal, augmentation of autoreactive B cells which release autoantigens to draining lymph nodes, molecular mimicry and infection-induced bystander immune activation products play a role in autoimmunization process or perpetuating autoimmune neuroinflammation. The review stresses the importance of early detection, clinical recognition, proper antibody testing and early therapy initiation as these disorders, regardless of a known or not trigger, are potentially treatable responding to systemic immunotherapy with intravenous steroids, IVIg, rituximab or even bortezomid.


Asunto(s)
Bortezomib/uso terapéutico , Encefalitis/inmunología , Encefalitis/terapia , Enfermedad de Hashimoto/inmunología , Enfermedad de Hashimoto/terapia , Inmunoglobulinas Intravenosas/uso terapéutico , Inmunoterapia , Rituximab/uso terapéutico , Esteroides/uso terapéutico , Autoanticuerpos/inmunología , Encefalitis/epidemiología , Encefalitis/patología , Enfermedad de Hashimoto/epidemiología , Enfermedad de Hashimoto/patología , Humanos , Incidencia , Sinapsis/inmunología
4.
BMC Neurol ; 19(1): 1, 2019 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-30606131

RESUMEN

BACKGROUND: Stiff Person Syndrome (SPS) is an under-diagnosed disorder that affects mobility and the quality of life of affected patients. The aim of the study is to describe the natural history of SPS, the extent of accumulated disability and the associated clinical and immunological features in patients followed for up to 8 years in a single center. METHODS: Our collective cohort included 57 SPS patients. Additionally, 32 of these patients were examined every 6 months for a two-year period in a longitudinal study protocol, to assess disease progression using quantitative measures of stiffness and heightened sensitivity. RESULTS: The most frequent initial symptom was leg stiffness, followed by paraspinal muscle rigidity and painful spasms in 95% of the patients. Although none of the patients required assistance for ambulation during the first 2 years of disease onset, 46 patients (80%) lost the ability to walk independently during our follow-up, despite symptomatic medications. In the longitudinal cohort, the number of stiff areas increased (p < 0.0001), consistent with worsening functional status and quality of life. High-titer anti-GAD antibodies were present in serum and CSF with elevated intrathecal GAD-specific IgG synthesis, but they did not correlate with clinical severity or progression. CONCLUSIONS: This large study on SPS patients, combining an eight-year follow-up at a single center by the same leading neurologist and his team, is the first to provide longitudinal data in a large patient subgroup using objective clinical measures. One of the main findings is that SPS is a progressive disease leading to physical disability over time.


Asunto(s)
Síndrome de la Persona Rígida , Estudios de Cohortes , Progresión de la Enfermedad , Humanos , Calidad de Vida
5.
Ann Neurol ; 82(2): 271-277, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28749549

RESUMEN

OBJECTIVE: In stiff person syndrome (SPS), an antibody-mediated impaired γ-aminobutyric acidergic (GABAergic) neurotransmission is believed to cause muscle stiffness and spasms. Most patients improve with GABA-enhancing drugs and intravenous immunoglobulin, but some respond poorly and remain disabled. The need for more effective therapy prompted a trial with the anti-CD20 monoclonal antibody rituximab. METHODS: This was a placebo-controlled randomized trial of rituximab (2 biweekly infusions of 1g each). The primary outcome was a change in stiffness scores at 6 months. Secondary outcomes were changes in heightened-sensitivity and quality of life scores. Enrolling 24 patients was calculated to detect 50% change in stiffness scores. RESULTS: Randomization was balanced for age, sex, disease duration, and glutamic acid decarboxylase autoantibody titers. No significant changes were noted at 6 months after treatment in all outcomes. Specifically, no differences were noted in the stiffness index, the primary outcome, or sensitivity scores, the secondary outcome, at 3 or 6 months. Quality of life scores improved significantly (p < 0.01) at 3 months in both groups, but not at 6 months, denoting an early placebo effect. Blinded self-assessment rating of the overall stiffness for individual patients revealed improvement in 4 patients in each group. At 6 months, improvement persisted in 1 patient in the placebo group versus 3 of 4 in the rituximab group, where these meaningful improvements were also captured by video recordings. INTERPRETATION: This is the largest controlled trial conducted in SPS patients and demonstrates no statistically significant difference in the efficacy measures between rituximab and placebo. Rituximab's lack of efficacy could be due to a considerable placebo effect; insensitivity of scales to quantify stiffness, especially in the less severely affected patients; or drug effectiveness in only a small patient subset. Ann Neurol 2017;82:271-277.


Asunto(s)
Rituximab/uso terapéutico , Síndrome de la Persona Rígida/tratamiento farmacológico , Autoanticuerpos/sangre , Método Doble Ciego , Femenino , Glutamato Descarboxilasa/inmunología , Humanos , Factores Inmunológicos/uso terapéutico , Masculino , Persona de Mediana Edad , Síndrome de la Persona Rígida/sangre , Resultado del Tratamiento
7.
Biochim Biophys Acta ; 1852(4): 658-66, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24949885

RESUMEN

Autoimmune neuropathies occur when immunologic tolerance to myelin or axonal antigens is lost. Even though the triggering factors and the underling immunopathology have not been fully elucidated in all neuropathy subsets, immunological studies on the patients' nerves, transfer experiments with the patients' serum or intraneural injections, and molecular fingerprinting on circulating autoantibodies or autoreactive T cells, indicate that cellular and humoral factors, either independently or in concert with each other, play a fundamental role in their cause. The review is focused on the main subtypes of autoimmune neuropathies, mainly the Guillain-Barré syndrome(s), the Chronic Inflammatory Demyelinating Polyneuropathy (CIDP), the Multifocal Motor Neuropathy (MMN), and the IgM anti-MAG-antibody mediated neuropathy. It addresses the factors associated with breaking tolerance, examines the T cell activation process including co-stimulatory molecules and key cytokines, and discusses the role of antibodies against peripheral nerve glycolipids or glycoproteins. Special attention is given to the newly identified proteins in the nodal, paranodal and juxtaparanodal regions as potential antigenic targets that could best explain conduction failure and rapid recovery. New biological agents against T cells, cytokines, B cells, transmigration and transduction molecules involved in their immunopathologic network, are discussed as future therapeutic options in difficult cases. This article is part of a Special Issue entitled: Neuromuscular Diseases: Pathology and Molecular Pathogenesis.


Asunto(s)
Síndrome de Guillain-Barré , Enfermedad de la Neurona Motora , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante , Autoanticuerpos/inmunología , Síndrome de Guillain-Barré/inmunología , Síndrome de Guillain-Barré/patología , Humanos , Enfermedad de la Neurona Motora/inmunología , Enfermedad de la Neurona Motora/patología , Proteínas del Tejido Nervioso/inmunología , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/inmunología , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/patología , Linfocitos T/inmunología , Linfocitos T/patología
8.
BMC Neurol ; 16: 48, 2016 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-27083892

RESUMEN

BACKGROUND: Mechanisms of inflammation and protein accumulation are crucial in inclusion body myositis (IBM). Recent evidence demonstrated that intravenous immunoglobulin failed to suppress cell-stress mediators in IBM. Here we studied the molecular changes in skeletal muscle biopsies from patients with IBM before and after treatment with alemtuzumab. METHODS: Relevant inflammatory and degeneration-associated markers were assessed by quantitative-PCR and immunohistochemistry in repeated muscle biopsy specimens from patients with IBM, which had been treated in a previously published uncontrolled proof-of-concept trial with alemtuzumab. RESULTS: There were no significant changes of the mRNA expression levels of the pro-inflammatory chemokines CXCL-9, CCL-4, and the cytokines IFN-γ, TGF-ß, TNF-α, and IL-1ß. Similarly, the degeneration-associated molecules ubiquitin, APP and αB-crystallin did not substantially change. Although no overall beneficial treatment effect was noted except for a 6-month stabilization, some patients experienced a transient improvement in muscle strength. In such responders, a trend towards reduced expression of inflammatory markers was noted. In contrast, the expression remained unchanged in the others who did not experience any change. The expression levels of IL-1ß and MHC-I correlated with the positive clinical effect. By immunohistochemistry, some inflammatory mediators like CD8, CXCL-9, and MHC-I were downmodulated. However, no consistent changes were noted for ubiquitin, nitrotyrosin and ß-amyloid. CONCLUSIONS: Alemtuzumab showed a trend towards downregulation of the expression of some inflammatory molecules in skeletal muscle of IBM patients but has no effect on several crucial markers of cell stress and degeneration. The data are helpful to explain the molecular treatment effects of future lymphocyte-targeted immunotherapies in IBM.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Inflamación/patología , Miositis por Cuerpos de Inclusión/tratamiento farmacológico , Alemtuzumab , Biopsia , Citocinas/metabolismo , Humanos , Inmunohistoquímica , Inmunoterapia/métodos , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/metabolismo
9.
Muscle Nerve ; 52(4): 498-502, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25728021

RESUMEN

INTRODUCTION: Electrodiagnostic studies (EDX) are not performed routinely before treatment suspension in CIDP, and no data exist regarding their value in predicting clinical relapse. METHODS: Serial EDX (baseline and after IGIV-C therapy) were analyzed from subjects in the ICE clinical trial who responded to IGIV-C treatment and were subsequently re-randomized to placebo in an extension phase. Comparisons were made between subjects who relapsed and those who did not. RESULTS: A total of 55% (6/11) of the Relapse group had an increase in total number of demyelinating findings (DF) versus 8% (1/13) in the No Relapse group (P = 0.023). In the Relapse group, 100% had ≥ 1 new DF and 73% (8/11) had ≥ 4 new DF versus 60% (8/13) and 8% (1/13), respectively, in the No Relapse group. CONCLUSIONS: An increased total number of DF or the occurrence of ≥ 4 new DF may indicate a higher risk of clinical relapse after treatment cessation in IGIV-C-responsive patients.


Asunto(s)
Inmunoglobulinas Intravenosas/uso terapéutico , Factores Inmunológicos/uso terapéutico , Conducción Nerviosa/efectos de los fármacos , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/terapia , Resultado del Tratamiento , Potenciales de Acción/efectos de los fármacos , Adulto , Anciano , Electrodiagnóstico , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Conducción Nerviosa/fisiología , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/fisiopatología , Tiempo de Reacción , Recurrencia
10.
Cochrane Database Syst Rev ; 7: CD001555, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-35658164

RESUMEN

BACKGROUND: Inclusion body myositis (IBM) is a late-onset inflammatory muscle disease (myopathy) associated with progressive proximal and distal limb muscle atrophy and weakness. Treatment options have attempted to target inflammatory and atrophic features of this condition (for example with immunosuppressive and immunomodulating drugs, anabolic steroids, and antioxidant treatments), although as yet there is no known effective treatment for reversing or minimising the progression of inclusion body myositis. In this review we have considered the benefits, adverse effects, and costs of treatment in targeting cardinal effects of the condition, namely muscle atrophy, weakness, and functional impairment. OBJECTIVES: To assess the effects of treatment for IBM. SEARCH METHODS: On 7 October 2014 we searched the Cochrane Neuromuscular Disease Group Specialized Register, the Cochrane Central Register for Controlled Trials (CENTRAL), MEDLINE, and EMBASE. Additionally in November 2014 we searched clinical trials registries for ongoing or completed but unpublished trials. SELECTION CRITERIA: We considered randomised or quasi-randomised trials, including cross-over trials, of treatment for IBM in adults compared to placebo or any other treatment for inclusion in the review. We specifically excluded people with familial IBM and hereditary inclusion body myopathy, but we included people who had connective tissue and autoimmune diseases associated with IBM, which may or may not be identified in trials. We did not include studies of exercise therapy or dysphagia management, which are topics of other Cochrane systematic reviews. DATA COLLECTION AND ANALYSIS: We used standard Cochrane methodological procedures. MAIN RESULTS: The review included 10 trials (249 participants) using different treatment regimens. Seven of the 10 trials assessed single agents, and 3 assessed combined agents. Many of the studies did not present adequate data for the reporting of the primary outcome of the review, which was the percentage change in muscle strength score at six months. Pooled data from two trials of interferon beta-1a (n = 58) identified no important difference in normalised manual muscle strength sum scores from baseline to six months (mean difference (MD) -0.06, 95% CI -0.15 to 0.03) between IFN beta-1a and placebo (moderate-quality evidence). A single trial of methotrexate (MTX) (n = 44) provided moderate-quality evidence that MTX did not arrest or slow disease progression, based on reported percentage change in manual muscle strength sum scores at 12 months. None of the fully published trials were adequately powered to detect a treatment effect. We assessed six of the nine fully published trials as providing very low-quality evidence in relation to the primary outcome measure. Three trials (n = 78) compared intravenous immunoglobulin (combined in one trial with prednisone) to a placebo, but we were unable to perform meta-analysis because of variations in study analysis and presentation of trial data, with no access to the primary data for re-analysis. Other comparisons were also reported in single trials. An open trial of anti-T lymphocyte immunoglobulin (ATG) combined with MTX versus MTX provided very low-quality evidence in favour of the combined therapy, based on percentage change in quantitative muscle strength sum scores at 12 months (MD 12.50%, 95% CI 2.43 to 22.57). Data from trials of oxandrolone versus placebo, azathioprine (AZA) combined with MTX versus MTX, and arimoclomol versus placebo did not allow us to report either normalised or percentage change in muscle strength sum scores. A complete analysis of the effects of arimoclomol is pending data publication. Studies of simvastatin and bimagrumab (BYM338) are ongoing. All analysed trials reported adverse events. Only 1 of the 10 trials interpreted these for statistical significance. None of the trials included prespecified criteria for significant adverse events. AUTHORS' CONCLUSIONS: Trials of interferon beta-1a and MTX provided moderate-quality evidence of having no effect on the progression of IBM. Overall trial design limitations including risk of bias, low numbers of participants, and short duration make it difficult to say whether or not any of the drug treatments included in this review were effective. An open trial of ATG combined with MTX versus MTX provided very low-quality evidence in favour of the combined therapy based on the percentage change data given. We were unable to draw conclusions from trials of IVIg, oxandrolone, and AZA plus MTX versus MTX. We need more randomised controlled trials that are larger, of longer duration, and that use fully validated, standardised, and responsive outcome measures.


ANTECEDENTES: La miositis por cuerpos de inclusión (MCI) es una enfermedad muscular inflamatoria (miopatía) de aparición tardía asociada con atrofia muscular y debilidad progresivas de los miembros proximales y distales. Las opciones de tratamiento se han intentado dirigir a las características inflamatorias y atróficas de esta afección (por ejemplo, con fármacos inmunosupresores e inmunomoduladores, esteroides anabólicos y tratamientos antioxidantes), aunque hasta ahora no hay un tratamiento eficaz conocido para la reversión o la reducción de la progresión de la miositis por cuerpos de inclusión. En esta revisión se han considerado los efectos beneficiosos, los efectos adversos y los costos del tratamiento dirigido a los efectos fundamentales de la afección, a saber, la atrofia muscular, la debilidad y el deterioro funcional. OBJETIVOS: Evaluar los efectos del tratamiento para la MCI. MÉTODOS DE BÚSQUEDA: El 7 octubre 2014, se hicieron búsquedas en el registro especializado del Grupo Cochrane de Enfermedades Neuromusculares (Cochrane Neuromuscular Disease Group), en el Registro Cochrane Central de Ensayos Controlados (Cochrane Central Register of Controlled Trials) (CENTRAL), MEDLINE y en EMBASE. Además, en noviembre 2014 se realizaron búsquedas de ensayos en curso o terminadas pero no publicados en los registros de ensayos clínicos. CRITERIOS DE SELECCIÓN: Se consideraron para inclusión en la revisión los ensayos aleatorios o cuasialeatorios, incluidos los ensayos cruzados (crossover), del tratamiento para la MCI en adultos en comparación con placebo u otro tratamiento. Se excluyeron específicamente los pacientes con MCI familiar y miopatía por cuerpos de inclusión hereditaria, pero se incluyeron los pacientes con enfermedades del tejido conjuntivo y autoinmunitarias asociadas con MCI, que pueden o no identificarse en los ensayos. No se incluyeron los estudios de terapia con ejercicios o tratamiento de la disfagia, que son los temas de otras revisiones sistemáticas Cochrane. OBTENCIÓN Y ANÁLISIS DE LOS DATOS: Se utilizaron procedimientos metodológicos Cochrane estándar. RESULTADOS PRINCIPALES: La revisión incluyó diez ensayos (249 participantes) que utilizaron diferentes regímenes de tratamiento. Siete de los diez ensayos evaluaron agentes únicos y tres evaluaron agentes combinados. Muchos de los estudios no presentaron datos suficientes para el informe del resultado primario de la revisión, que fue el cambio porcentual en la puntuación de fuerza muscular a los seis meses. Los datos agrupados de dos ensayos de interferón beta­1a (n = 58) no identificaron diferencias importantes en las puntuaciones normalizadas de la suma de la fuerza muscular manual desde el inicio hasta los seis meses (diferencia de medias [DM] ­0,06; IC del 95%: ­0,15 a 0,03) entre IFN beta­1a y placebo (pruebas de calidad moderada). Un único ensayo de metotrexato (MTX) (n = 44) proporcionó pruebas de calidad moderada de que el MTX no detuvo ni enlenteció la progresión de la enfermedad, sobre la base del cambio porcentual informado en las puntuaciones de la suma de la fuerza muscular manual a los 12 meses. Ninguno de los ensayos publicados completamente tuvo poder estadístico suficiente para detectar un efecto del tratamiento. Se consideró que seis de los nueve ensayos publicados completamente aportaron pruebas de calidad muy baja con respecto a la medida de resultado primaria. Tres ensayos (n = 78) compararon la inmunoglobulina intravenosa (combinada en un ensayo con prednisona) con placebo, pero no fue posible realizar el metanálisis debido a las variaciones en el análisis de los estudios y a la presentación de los datos del ensayo, sin acceso a los datos primarios para el reanálisis. Otras comparaciones también se informaron en ensayos individuales. Un ensayo abierto de inmunoglobulina anti­linfocitos T (IgAT) combinada con MTX versus MTX proporcionó pruebas de calidad muy baja a favor del tratamiento combinado, sobre la base del cambio porcentual en las puntuaciones cuantitativas de la suma de la fuerza muscular a los 12 meses (DM 12,50%; IC del 95%: 2,43 a 22,57). Los datos de los ensayos de oxandrolona versus placebo, azatioprina (AZA) combinada con MTX versus MTX y arimoclomol versus placebo no permitieron informar sobre el cambio porcentual o normalizado en las puntuaciones de la suma de la fuerza muscular. Un análisis completo de los efectos del arimoclomol está pendiente de la publicación de los datos. Están en curso estudios de simvastatina y bimagrumab (BYM338). Todos los ensayos analizados informaron eventos adversos. Solamente uno de los diez ensayos interpretó la significación estadística de los eventos adversos. Ninguno de los ensayos incluyó criterios preespecificados para los eventos adversos significativos. CONCLUSIONES DE LOS AUTORES: Los ensayos de interferón beta­1a y MTX proporcionaron pruebas de calidad moderada de que no tienen efectos sobre la progresión de la MCI. Las limitaciones generales del diseño de los ensayos, que incluyen el riesgo de sesgo, los escasos números de participantes y la corta duración, hacen difícil determinar si alguno de los tratamientos farmacológicos incluidos en esta revisión fue eficaz. Un ensayo abierto de ATG combinada con MTX versus MTX aportó pruebas de calidad muy baja a favor del tratamiento combinado sobre la base de los datos de cambio porcentual facilitados. No fue posible establecer conclusiones de los ensayos de IgIV, oxandrolona y AZA más MTX versus MTX. Se necesitan más ensayos controlados aleatorios de mayor tamaño, con una duración más prolongada y que utilicen medidas de resultado completamente validadas, estandarizadas y de interés.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA