Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Brain ; 143(3): 960-975, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32203581

RESUMEN

We performed post-mortem studies on two patients with advanced Parkinson's disease 8 and10 years following AAV2-neurturin (CERE120) gene therapy, the longest post-mortem trophic factor gene therapy cases reported to date. CERE120 was delivered to the putamen bilaterally in one case (10 years post-surgery), and to the putamen plus the substantia nigra bilaterally in the second (8 years post-surgery). In both patients there was persistent, albeit limited, neurturin expression in the putamen covering ∼3-12% of the putamen. In the putamen, dense staining of tyrosine hydroxylase-positive fibres was observed in areas that contained detectable neurturin expression. In the substantia nigra, neurturin expression was detected in 9.8-18.95% and 22.02-39% of remaining melanin-containing neurons in the patient with putamenal and combined putamenal and nigral gene delivery, respectively. Melanized neurons displayed intense tyrosine hydroxylase and RET proto-oncogene expression in nigral neurons in the patient where CERE120 was directly delivered to the nigra. There was no difference in the degree of Lewy pathology in comparison to untreated control patients with Parkinson's disease, and α-synuclein aggregates were detected in neurons that also stained for neurturin, RET, and tyrosine hydroxylase. These changes were not associated with antiparkinsonian benefits likely due to the limited neurturin expression. This study provides the longest term evidence of persistent transgene expression following gene delivery to the CNS and the first human results when targeting both the terminal fields in the putamen as well as the originating nigral neurons.


Asunto(s)
Terapia Genética , Neurturina/biosíntesis , Enfermedad de Parkinson/metabolismo , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Humanos , Cuerpos de Lewy/metabolismo , Melaninas/inmunología , Persona de Mediana Edad , Neuronas/inmunología , Neurturina/administración & dosificación , Enfermedad de Parkinson/inmunología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-ret/biosíntesis , Putamen/inmunología , Putamen/metabolismo , Sustancia Negra/inmunología , Sustancia Negra/metabolismo , Tirosina 3-Monooxigenasa/inmunología , alfa-Sinucleína/metabolismo
2.
Neurobiol Dis ; 97(Pt B): 169-178, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27063797

RESUMEN

The therapeutic potential of neurotrophic factors has been recognized for decades, with clinical trials in human neurodegenerative diseases extending back at least 25years. While improvements in clinical dosing paradigms have reduced the side effects commonly seen in the earlier trials, efficacy has remained a serious disappointment (reviewed in Bartus and Johnson, 2016). This lengthy clinical effort stands in contrast to robust effects consistently achieved from different neurotrophic factors in a variety of animal models of neurodegeneration. This review discusses the prevailing assumption and supporting data that the major reason for the disappointing efficacy of past clinical trials is related to suboptimal dosing methods. It is concluded that while further improvements in dosing parameters might be useful, a much greater problem centers around a number of specific morphologic and functional changes in neurons in human neurodegenerative disease that mitigate the ability of neurotrophic factors to exert their effects. Moreover, the biological substrate which neurotrophic factors depend upon to exert their effects continues to erode as time progresses, due to the progressive nature of these diseases. For this reason, most of the empirically-supported reasons contributing to the weak neurotrophic responses in human patients can be mitigated by enrolling less severely advanced cases. It is further concluded that recent clinical trials of neurotrophic factors have generated important evidence that shifts risk: benefit assessments to support enrolling earlier-stage patients. While the Alzheimer's field has begun to shift attention toward much earlier-stage (even prodromal) patients in trials intended to modify disease progression, other neurodegenerative diseases (e.g., Parkinson's, ALS and possibly HD) must now consider similar changes in approach.


Asunto(s)
Factores de Crecimiento Nervioso/administración & dosificación , Enfermedades Neurodegenerativas/terapia , Fármacos Neuroprotectores/administración & dosificación , Animales , Ensayos Clínicos como Asunto , Humanos , Factores de Crecimiento Nervioso/genética
3.
Neurobiol Dis ; 97(Pt B): 156-168, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27063798

RESUMEN

Over the past 25years, about 3 dozen clinical reports have been published regarding the safety and possible efficacy of neurotrophic factors in patients with various neurodegenerative diseases. This effort involved a half dozen different neurotrophic factors, using at least 5 different general delivery approaches for ALS (amyolateral sclerosis), peripheral neuropathies, PD (Parkinson's disease) and AD (Alzheimer's disease). While none of these efforts have yet produced efficacy data sufficiently robust or reliable to establish neurotrophic factors as treatments for any human disease, the obstacles encountered and novel information reported, when viewed collectively, provide important insight to help future efforts. Three consistent themes emerge from these publications: (1) unexpected and undesirable side effects, at times serious, have plagued many efforts to deliver neurotrophic factors to humans; (2) the magnitude and consistency of clinical benefit has been disappointing; (3) by far that most consistently proposed reason for the side effects and poor efficacy has been inadequate dosing and delivery. This paper reviews and attempts to synthesize the available data derived from clinical tests of neurotrophic factors for neurodegenerative diseases. The obstacles encountered, the solutions attempted, and the lessons learned are discussed. The vast majority of solutions have involved changes in dosing paradigms and dose levels, which has primarily led to improved safety outcomes. However, lack of adequate efficacy remains a significant issue. While current efforts continue to focus exclusively on still-further changes in dosing parameters, a review of available data argues that it may now be the time to ask whether other, non-dose-related variables should be given more serious consideration as being responsible for the great divide that exists between the robust effects seen in animal models and the relatively weak effects seen in human neurodegenerative patients. Foremost among these appears to be the severe degeneration seen in the majority of patients enrolled in past and current trials testing neurotrophic factors in humans. A companion paper (Bartus and Johnson, 2016), reviews the contemporary data and concludes that compelling empirical evidence already exists for enrolling earlier-stage subjects as likely essential to achieving more robust and reliable benefit.


Asunto(s)
Factores de Crecimiento Nervioso/administración & dosificación , Enfermedades Neurodegenerativas/terapia , Fármacos Neuroprotectores/administración & dosificación , Animales , Ensayos Clínicos como Asunto , Humanos , Factores de Crecimiento Nervioso/genética
4.
Neurobiol Dis ; 85: 11-24, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26459114

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a chronic and progressive neuromuscular disease for which no cure exists and better treatment options are desperately needed. We hypothesize that currently approved ß2-adrenoceptor agonists may effectively treat the symptoms and possibly slow the progression of ALS. Although ß2-agonists are primarily used to treat asthma, pharmacologic data from animal models of neuromuscular diseases suggest that these agents may have pharmacologic effects of benefit in treating ALS. These include inhibiting protein degradation, stimulating protein synthesis, inducing neurotrophic factor synthesis and release, positively modulating microglial and systemic immune function, maintaining the structural and functional integrity of motor endplates, and improving energy metabolism. Moreover, stimulation of ß2-adrenoceptors can activate a range of downstream signaling events in many different cell types that could account for the diverse array of effects of these agents. The evidence supporting the possible therapeutic benefits of ß2-agonists is briefly reviewed, followed by a more detailed review of clinical trials testing the efficacy of ß-agonists in a variety of human neuromuscular maladies. The weight of evidence of the potential benefits from treating these diseases supports the hypothesis that ß2-agonists may be efficacious in ALS. Finally, ways to monitor and manage the side effects that may arise with chronic administration of ß2-agonists are evaluated. In sum, effective, safe and orally-active ß2-agonists may provide a novel and convenient means to reduce the symptoms of ALS and possibly delay disease progression, affording a unique opportunity to repurpose these approved drugs for treating ALS, and rapidly transforming the management of this serious, unmet medical need.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/administración & dosificación , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Fármacos Neuroprotectores/administración & dosificación , Administración Oral , Agonistas de Receptores Adrenérgicos beta 2/efectos adversos , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Humanos , Fármacos Neuroprotectores/efectos adversos
5.
Ann Neurol ; 78(2): 248-57, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26061140

RESUMEN

OBJECTIVE: A 12-month double-blind sham-surgery-controlled trial assessing adeno-associated virus type 2 (AAV2)-neurturin injected into the putamen bilaterally failed to meet its primary endpoint, but showed positive results for the primary endpoint in the subgroup of subjects followed for 18 months and for several secondary endpoints. Analysis of postmortem tissue suggested impaired axonal transport of neurturin from putamen to substantia nigra. In the present study, we tested the safety and efficacy of AAV2-neurturin delivered to putamen and substantia nigra. METHODS: We performed a 15- to 24-month, multicenter, double-blind trial in patients with advanced Parkinson disease (PD) who were randomly assigned to receive bilateral AAV2-neurturin injected bilaterally into the substantia nigra (2.0 × 10(11) vector genomes) and putamen (1.0 × 10(12) vector genomes) or sham surgery. The primary endpoint was change from baseline to final visit performed at the time the last enrolled subject completed the 15-month evaluation in the motor subscore of the Unified Parkinson's Disease Rating Scale in the practically defined off state. RESULTS: Fifty-one patients were enrolled in the trial. There was no significant difference between groups in the primary endpoint (change from baseline: AAV2-neurturin, -7.0 ± 9.92; sham, -5.2 ± 10.01; p = 0.515) or in most secondary endpoints. Two subjects had cerebral hemorrhages with transient symptoms. No clinically meaningful adverse events were attributed to AAV2-neurturin. INTERPRETATION: AAV2-neurturin delivery to the putamen and substantia nigra bilaterally in PD was not superior to sham surgery. The procedure was well tolerated, and there were no clinically significant adverse events related to AAV2-neurturin.


Asunto(s)
Transporte Axonal , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Neurturina/genética , Enfermedad de Parkinson/terapia , Putamen/metabolismo , Sustancia Negra/metabolismo , Anciano , Dependovirus , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/fisiopatología , Putamen/fisiopatología , Sustancia Negra/fisiopatología , Resultado del Tratamiento
6.
Mov Disord ; 30(13): 1715-24, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26769457

RESUMEN

Trophic factors show great promise in laboratory studies as potential therapies for PD. However, multiple double-blind, clinical trials have failed to show benefits in comparison to a placebo control. This article will review the scientific rationale for testing trophic factors in PD, the results of the different clinical trials that have been performed to date, and the possible explanations for these failed outcomes. We will also consider future directions and the likelihood that trophic factors will become a viable therapy for patients with PD.


Asunto(s)
Antiparkinsonianos/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular , Enfermedad de Parkinson/terapia , Animales , Antiparkinsonianos/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico
7.
Mol Ther ; 22(3): 487-497, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24356252

RESUMEN

Over the past decade, nine gene therapy clinical trials for Parkinson's disease (PD) have been initiated and completed. Starting with considerable optimism at the initiation of each trial, none of the programs has yet borne sufficiently robust clinical efficacy or found a clear path toward regulatory approval. Despite the immediately disappointing nature of the efficacy outcomes in these trials, the clinical data garnered from the individual studies nonetheless represent tangible and significant progress for the gene therapy field. Collectively, the clinical trials demonstrate that we have overcome the major safety hurdles previously suppressing central nervous system (CNS) gene therapy, for none produced any evidence of untoward risk or harm after administration of various vector-delivery systems. More importantly, these studies also demonstrated controlled, highly persistent generation of biologically active proteins targeted to structures deep in the human brain. Therefore, a renewed, focused emphasis must be placed on advancing clinical efficacy by improving clinical trial design, patient selection and outcome measures, developing more predictive animal models to support clinical testing, carefully performing retrospective analyses, and most importantly moving forward-beyond our past limits.


Asunto(s)
Sistema Nervioso Central/metabolismo , Terapia Genética/métodos , Enfermedad de Parkinson/terapia , Animales , Sistema Nervioso Central/patología , Ensayos Clínicos como Asunto , Terapia Genética/efectos adversos , Vectores Genéticos/uso terapéutico , Humanos , Enfermedad de Parkinson/genética , Proyectos de Investigación
8.
Brain ; 136(Pt 8): 2419-31, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23884810

RESUMEN

The pace of nigrostriatal degeneration, both with regards to striatal denervation and loss of melanin and tyrosine hydroxylase-positive neurons, is poorly understood especially early in the Parkinson's disease process. This study investigated the extent of nigrostriatal degeneration in patients with Parkinson's disease at different disease durations from time of diagnosis. Brains of patients with Parkinson's disease (n=28) with post-diagnostic intervals of 1-27 years and normal elderly control subjects (n=9) were examined. Sections of the post-commissural putamen and substantia nigra pars compacta were processed for tyrosine hydroxylase and dopamine transporter immunohistochemistry. The post-commissural putamen was selected due to tissue availability and the fact that dopamine loss in this region is associated with motor disability in Parkinson's disease. Quantitative assessments of putaminal dopaminergic fibre density and stereological estimates of the number of melanin-containing and tyrosine hydroxylase-immunoreactive neurons in the substantia nigra pars compacta (both in total and in subregions) were performed by blinded investigators in cases where suitable material was available (n=17). Dopaminergic markers in the dorsal putamen showed a modest loss at 1 year after diagnosis in the single case available for study. There was variable (moderate to marked) loss, at 3 years. At 4 years post-diagnosis and thereafter, there was virtually complete loss of staining in the dorsal putamen with only an occasional abnormal dopaminergic fibre detected. In the substantia nigra pars compacta, there was a 50-90% loss of tyrosine hydroxylase-positive neurons from the earliest time points studied with only marginal additional loss thereafter. There was only a ∼10% loss of melanized neurons in the one case evaluated 1 year post-diagnosis, and variable (30 to 60%) loss during the first several years post-diagnosis with more gradual and subtle loss in the second decade. At all time points, there were more melanin-containing than tyrosine hydroxylase-positive cells. Loss of dopaminergic markers in the dorsal putamen occurs rapidly and is virtually complete by 4 years post-diagnosis. Loss of melanized nigral neurons lags behind the loss of dopamine markers. These findings have important implications for understanding the nature of Parkinson's disease neurodegeneration and for studies of putative neuroprotective/restorative therapies.


Asunto(s)
Cuerpo Estriado/patología , Neuronas/patología , Enfermedad de Parkinson/patología , Sustancia Negra/patología , Adulto , Anciano , Anciano de 80 o más Años , Cuerpo Estriado/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Sustancia Negra/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
9.
Alzheimers Dement ; 10(5): 571-81, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24411134

RESUMEN

BACKGROUND: Nerve growth factor (NGF) is an endogenous neurotrophic-factor protein with the potential to restore function and to protect degenerating cholinergic neurons in Alzheimer's disease (AD), but safe and effective delivery has proved unsuccessful. METHODS: Gene transfer, combined with stereotactic surgery, offers a potential means to solve the long-standing delivery obstacles. An open-label clinical trial evaluated the safety and tolerability, and initial efficacy of three ascending doses of the genetically engineered gene-therapy vector adeno-associated virus serotype 2 delivering NGF (AAV2-NGF [CERE-110]). Ten subjects with AD received bilateral AAV2-NGF stereotactically into the nucleus basalis of Meynert. RESULTS: AAV2-NGF was safe and well-tolerated for 2 years. Positron emission tomographic imaging and neuropsychological testing showed no evidence of accelerated decline. Brain autopsy tissue confirmed long-term, targeted, gene-mediated NGF expression and bioactivity. CONCLUSIONS: This trial provides important evidence that bilateral stereotactic administration of AAV2-NGF to the nucleus basalis of Meynert is feasible, well-tolerated, and able to produce long-term, biologically active NGF expression, supporting the initiation of an ongoing multicenter, double-blind, sham-surgery-controlled trial.


Asunto(s)
Enfermedad de Alzheimer/terapia , Dependovirus/genética , Terapia Genética/métodos , Factor de Crecimiento Nervioso/genética , Anciano , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Núcleo Basal de Meynert , Estudios de Factibilidad , Femenino , Vectores Genéticos , Humanos , Inmunohistoquímica , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Pruebas Neuropsicológicas , Procedimientos Neuroquirúrgicos , Tomografía de Emisión de Positrones , Técnicas Estereotáxicas , Resultado del Tratamiento
10.
Neurobiol Dis ; 58: 38-48, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23631873

RESUMEN

This paper reassesses the currently accepted viewpoint that targeting the terminal fields (i.e. striatum) of degenerating nigrostriatal dopamine neurons with neurotrophic factors in Parkinson's disease (PD) is sufficient for achieving an optimal neurotrophic response. Recent insight indicating that PD is an axonopathy characterized by axonal transport deficits prompted this effort. We tested whether a significantly greater neurotrophic response might be induced in SN neurons when the neurotrophic factor neurturin (NRTN) is also targeted to the substantia nigra (SN), compared to the more conventional, striatum-only target. While recognizing the importance of maintaining the integrity of nigrostriatal fibers and terminals (especially for achieving optimal function), we refocused attention to the fate of SN neurons. Under conditions of axonal degeneration and neuronal transport deficits, this component of the nigrostriatal system is most vulnerable to the lack of neurotrophic exposure following striatal-only delivery. Given the location of repair genes induced by neurotrophic factors, achieving adequate neurotrophic exposure to the SN neurons is essential for an optimal neurotrophic response, while the survival of these neurons is essential to the very survival of the fibers. Two separate studies were performed using the 6-OHDA model of nigrostriatal degeneration, in conjunction with delivery of the viral vector AAV2-NRTN (CERE-120) to continuously express NRTN to either striatum or nigra alone or combined striatal/nigral exposure, including conditions of ongoing axonopathy. These studies provide additional insight for reinterpreting past animal neurotrophic/6-OHDA studies conducted under conditions where axon transport deficiencies were generally not accounted for, which suggested that targeting the striatum was both necessary and sufficient. The current data demonstrate that delivering NRTN directly to the SN produces 1) expanded NRTN distribution within the terminal field and cell bodies of targeted nigrostriatal neurons, 2) enhanced intracellular neurotrophic factor signaling in the nigrostriatal neurons, and 3) produced greater numbers of surviving dopamine neurons against 6-OHDA-induced toxicity, particularly under the conditions of active axonopathy. Thus, these data provide empirical support that targeting the SN with neurotrophic factors (in addition to striatum) may help enhance the neurotrophic response in midbrain neurons, particularly under conditions of active neurodegeneration which occurs in PD patients.


Asunto(s)
Adenoviridae/genética , Cuerpo Estriado/metabolismo , Factores de Crecimiento Nervioso/administración & dosificación , Enfermedades Neurodegenerativas/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Sustancia Negra/metabolismo , Adrenérgicos/toxicidad , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Vectores Genéticos/fisiología , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/genética , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/patología , Fármacos Neuroprotectores/metabolismo , Oxidopamina/toxicidad , Ratas , Ratas Sprague-Dawley , Tirosina 3-Monooxigenasa
11.
Neurobiol Dis ; 48(2): 153-78, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22525569

RESUMEN

While the therapeutic potential of neurotrophic factors has been well-recognized for over two decades, attempts to translate that potential to the clinic have been disappointing, largely due to significant delivery obstacles. Similarly, gene therapy (or gene transfer) emerged as a potentially powerful, new therapeutic approach nearly two decades ago and despite its promise, also suffered serious setbacks when applied to the human clinic. As advances continue to be made in both fields, ironically, they may now be poised to complement each other to produce a translational breakthrough. The accumulated data argue that gene transfer provides the 'enabling technology' that can solve the age-old delivery problems that have plagued the translation of neurotrophic factors as treatments for chronic central nervous system diseases. A leading translational program applying gene transfer to deliver a neurotrophic factor to rejuvenate and protect degenerating human neurons is CERE-120 (AAV2-NRTN). To date, over two dozen nonclinical studies and three clinical trials have been completed. A fourth (pivotal) clinical trial has completed all dosing and is currently evaluating safety and efficacy. In total, eighty Parkinson's disease (PD) subjects have thus far been dosed with CERE-120 (some 7 years ago), representing over 250 cumulative patient-years of exposure, with no serious safety issues identified. In a completed sham-surgery, double-blinded controlled trial, though the primary endpoint (the Unified Parkinson's Disease Rating Scale (UDPRS) motor off score measured at 12 months) did not show benefit from CERE-120, several important motor and quality of life measurements did, including the same UPDRS-motor-off score, pre-specified to also be measured at a longer, 18-month post-dosing time point. Importantly, not a single measurement favored the sham control group. This study therefore, provided important, well-controlled evidence establishing 'clinical proof of concept' for gene transfer to the CNS and the first controlled evidence for clinical benefit of a neurotrophic factor in a human neurodegenerative disease. This paper reviews the development of CERE-120, starting historically with the long-standing interest in the therapeutic potential of neurotrophic factors and continuing with selective accounts of past efforts to translate their potential to the clinic, eventually leading to the application of gene transfer and its role as the 'enabling technology'. Because of growing interest in translational R&D, including its practice in industry, the paper is uniquely oriented from the author's personal, quasi-autobiographic perspective and career-long experiences conducting translational research and development, with a focus on various translational neurotrophic factor programs spanning 30+ years in Big Pharma and development-stage biotech companies. It is hoped that by sharing these perspectives, practical insight and information might be provided to others also interested in translational R&D as well as neurotrophic factors and gene therapy, offering readers the opportunity to benefit from some of our successes, while possibly avoiding some of our missteps.


Asunto(s)
Factores de Crecimiento Nervioso/uso terapéutico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Investigación Biomédica Traslacional , Animales , Biotecnología , Sistemas de Liberación de Medicamentos , Industria Farmacéutica , Terapia Genética/historia , Vectores Genéticos , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Factores de Crecimiento Nervioso/genética , Enfermedades del Sistema Nervioso/genética , Investigación Biomédica Traslacional/historia
12.
Neurobiol Dis ; 44(1): 38-52, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21704161

RESUMEN

Recent analyses of autopsied brains from subjects previously administered AAV2-neurturin (NRTN) gene transfer argues that optimizing the effects of neurotrophic factors in Parkinson's disease (PD) likely requires delivery to both the degenerating cell bodies (in substantia nigra) and their terminals (in striatum). Prior to implementing this novel dosing paradigm in humans, we conducted eight nonclinical experiments with three general objectives: (1) evaluate the feasibility, safety and effectiveness of targeting the substantia nigra (SN) with AAV2-NRTN, (2) better understand and appraise recent warnings of serious weight loss that might occur with targeting the SN with neurotrophic factors, and (3) define an appropriate dose of AAV2-NRTN that should safely and effectively cover the SN in PD patients. Toward these ends, we first determined SN volume for rats, monkeys and humans, and employed these values to calculate comparable dose equivalents for each species by scaling each dose, based on relative SN volume. Using this information, we next injected AAV2-GFP to monkey SN to quantify AAV2-vector distribution and confirm reasonable SN coverage. We then selected and administered a ~200-fold range of AAV2-NRTN doses (and a single AAV2-GDNF dose) to rat SN, producing a wide range of protein expression. In contrast to recent warnings regarding nigra targeting, no dose produced any serious side effects or toxicity, though we replicated the modest reduction in weight gain reported by others with the highest AAV2-NRTN and the AAV2-GDNF dose. A dose-related increase in NRTN expression was seen, with the lower doses limiting NRTN to the peri-SN and the highest dose producing mistargeted NRTN well outside the SN. We then demonstrated that the reduction in weight gain following excessive-doses can be dissociated from NRTN in the targeted SN, and is linked to mistargeted NRTN in the diencephalon. We also showed that prior destruction of the dopaminergic SN neurons via 6-OHDA had no impact on the weight loss phenomenon, further dissociating neurotrophic exposure to the SN as the culprit for weight changes. Finally, low AAV2-NRTN doses provided significant neuroprotection against 6-OHDA toxicity, establishing a wide therapeutic index for nigral targeting. These data support targeting the SN with AAV2-NRTN in PD patients, demonstrating that properly targeted and scaled AAV2-NRTN provides safe and effective NRTN expression. They also provided the means to define an appropriate human-equivalent dose for proceeding into an ongoing clinical trial, using empirically-based scaling to account for marked differences in SN volume between species.


Asunto(s)
Dependovirus/genética , Terapia Genética/métodos , Neurturina/metabolismo , Enfermedad de Parkinson/terapia , Sustancia Negra/metabolismo , Animales , Conducta Animal/fisiología , Dieta , Dosificación de Gen , Marcación de Gen , Vectores Genéticos , Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Proteínas Fluorescentes Verdes , Inmunohistoquímica , Masculino , Neuronas/metabolismo , Neurturina/efectos adversos , Ratas , Ratas Sprague-Dawley , Medición de Riesgo , Tirosina 3-Monooxigenasa/metabolismo , Aumento de Peso/genética , Aumento de Peso/fisiología , Pérdida de Peso/fisiología
13.
Mov Disord ; 26(1): 27-36, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21322017

RESUMEN

BACKGROUND: AAV2-neurturin (CERE-120) is designed to deliver the neurotrophic-factor, neurturin, to the striatum to restore and protect degenerating nigrostriatal neurons in Parkinson's disease (PD). A common hypothesis is that following expression in the striatum, neurotrophic-factors like neurturin (NRTN) will be transported from degenerating terminals to their cell bodies in the substantia nigra pars compacta (SNc). METHODS: We tested this concept using immunohistochemistry, comparing the bioactivity of AAV2-neurturin in brains of PD patients versus those of nonhuman primates similarly treated. RESULTS: NRTN-immunostaining in the targeted striatum was seen in all PD cases (mean putaminal coverage: ∼15% by volume); comparable expression was observed in young, aged, and parkinsonian monkeys. In the SNc cell bodies, however, only rare evidence of neurturin was seen in PD, while ample evidence of intense nigral-NRTN was observed in all monkeys. NRTN-expression was associated with occasional, sparse TH-induction in the striatum of PD, but nothing apparent in the SNc. In primates, NRTN produced robust TH-induction throughout the nigrostriatal neurons. DISCUSSION: These data provide the first evidence that gene therapy can increase expression of a neurotrophic-factor deep in the PD brain and that clear but modest enhancement of degenerating neurons can be induced. They also provide important insight regarding deficiencies in the status of nigrostriatal neurons in advanced PD, suggesting that serious axon-transport deficits reduced the bioactivity of AAV2-NRTN by limiting the protein exposed to the cell body. Thus, future efforts using neurotrophic-factors to treat neurodegenerative diseases will need to target both the terminal fields and the cell bodies of degenerating neurons to assure maximal benefit is achieved.


Asunto(s)
Cuerpo Estriado/metabolismo , Terapia Genética/métodos , Intoxicación por MPTP/terapia , Neurturina/uso terapéutico , Enfermedad de Parkinson/terapia , Anciano , Animales , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Lateralidad Funcional , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Intoxicación por MPTP/inducido químicamente , Intoxicación por MPTP/metabolismo , Intoxicación por MPTP/patología , Macaca mulatta , Masculino , Persona de Mediana Edad , Neurturina/genética , Neurturina/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Tirosina 3-Monooxigenasa/metabolismo
14.
Exp Neurol ; 323: 113091, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31678350

RESUMEN

Neurotrophic factors as candidates for ALS therapeutics have previously been studied in the context of attempts to slow disease progression. For a variety of reasons, clinical trials of neurotrophic factors have failed to show efficacy in ALS patients. Previous studies in Parkinson's Disease (PD) models have shown promise with the use of recombinant adeno-associated virus serotype-2 (rAAV2)-neurturin (NRTN) [AAV2-NRTN] providing neuroprotection and behavioral improvements in preclinical models which subsequently resulted in several clinical studies in patients with PD. Given that this neurotrophic compound has not been studied in the context of ALS, we conducted a study of AAV2-NRTN to assess the preclinical safety, tolerability, biodistribution, and efficacy of this compound in an ALS mouse model. SOD1G93A mice were injected with AAV2-NRTN intraspinally at several doses into the cervical spinal cord at 60 days of age. NRTN expression was noted in motor neurons (MNs) of the targeted cervical spinal cord as well as in their neuromuscular junction projections but not in the lumbar spinal cord, which was not targeted. Neuropathologically, a dose-dependent neuroprotective effect was seen in cervical MNs and neuromuscular junctions that was reflected in a slowing of forelimb grip strength decline. As expected, this neuroprotection was found to be focal and was not seen beyond the immediate region of injection. Overall, there were no increases in morbidity, changes in serum chemistries or blood counts and no cases of drug-related mortality. Because there is a broad clinical experience for this compound, these data provide evidence to support further investigation of AAV2-NRTN as a potential ALS therapeutic.


Asunto(s)
Esclerosis Amiotrófica Lateral , Técnicas de Transferencia de Gen , Neuronas Motoras/metabolismo , Neurturina/administración & dosificación , Animales , Médula Cervical/metabolismo , Dependovirus , Modelos Animales de Enfermedad , Vectores Genéticos , Humanos , Ratones , Fármacos Neuroprotectores/farmacología , Parvovirinae , Transducción Genética
15.
Neurobiol Dis ; 34(1): 40-50, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19150499

RESUMEN

Members of the GDNF family of ligands, including neurturin (NTN), have been implicated as potential therapeutic agents for Huntington's disease (HD). The present study examined the ability of CERE-120 (AAV2-NTN) to provide structural and functional protection in the N171-82Q transgenic HD mouse model. AAV2-NTN therapy attenuated rotorod deficits in this mutant relative to control treated transgenics (p<0.01). AAV2-NTN treatment significantly reduced the number of transgenic mice that exhibited clasping behavior and partially restored their stride lengths (both p<0.05). Stereological counts of NeuN-ir neurons revealed a significant neuroprotection in the striatum of AAV2-NTN treated relative to control treated transgenics (p<0.001). Most fascinating, stereological counts of NeuN-labeled cells in layers V-VI of prefrontal cortex revealed that intrastriatal AAV2-NTN administration prevented the loss of frontal cortical NeuN-ir neurons seen in transgenic mice (p<0.01). These data indicate that gene delivery of NTN may be a viable strategy for the treatment of this incurable disease.


Asunto(s)
Corteza Cerebral/fisiopatología , Cuerpo Estriado/fisiopatología , Terapia Genética , Enfermedad de Huntington/terapia , Actividad Motora , Neuronas/fisiología , Neurturina/genética , Animales , Proteínas de Unión al ADN , Dependovirus/genética , Modelos Animales de Enfermedad , Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Ratones , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neurturina/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Distribución Aleatoria , Prueba de Desempeño de Rotación con Aceleración Constante
16.
Neurobiol Dis ; 33(3): 473-81, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19135533

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a devastating disease that is characterized by the progressive loss of motor neurons. Patients with ALS usually die from respiratory failure due to respiratory muscle paralysis. Consequently, therapies aimed at preserving segmental function of the respiratory motor neurons could extend life for these patients. Insulin-like growth factor-I (IGF-I) is known to be a potent survival factor for motor neurons. In this study we induced high levels of IGF-I expression in the cervical spinal cord of hSOD1(G93A) rats with intraspinal cord (ISC) injections of an adeno-associated virus serotype 2 vector (CERE-130). This approach reduced the extent of motor neuron loss in the treated segments of the spinal cord. However, a corresponding preservation of motor function was observed in male, but not female, hSOD1(G93A) rats. We conclude that ISC injection of CERE-130 has the potential to protect motor neurons and preserve neuromuscular function in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Dependovirus/genética , Terapia Genética , Factor I del Crecimiento Similar a la Insulina/genética , Médula Espinal/metabolismo , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Supervivencia Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Vectores Genéticos , Inyecciones Espinales , Masculino , Actividad Motora , Neuronas Motoras/fisiología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Factores Sexuales , Análisis de Supervivencia , Transducción Genética
17.
Mol Ther ; 16(10): 1737-44, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18728639

RESUMEN

Neurturin (NTN) is a neurotrophic factor for dopaminergic neurons that may be therapeutic for patients with Parkinson's disease (PD). As a crucial component in a series of nonclinical translational studies aimed at testing whether CERE-120 should advance into clinical trials in PD subjects, we characterized the expression, bioactivity and safety of CERE-120, an adeno-associated virus type-2 (AAV2) vector encoding NTN, following delivery to the striatum of nonhuman primates. Monkeys received bilateral injections of CERE-120 across a tenfold range of doses (6 x 10(10) to 6 x 10(11) vector genomes per animal) or formulation buffer (FB) control. We report here, for the first time, a dose-related: increase in NTN protein expression within the striatum and substantia nigra (SN) pars compacta of nonhuman primates; increase in nigrostriatal tyrosine hydroxylase (TH), (the rate-limited enzyme for dopamine); and activation of phosphorylated signal-regulated kinase (a common neurotrophic signaling event). Additionally, extensive toxicology testing revealed no adverse effects of CERE-120 on in-life measures, neurotoxicity (in any site throughout the brain) or systemic pathology (in any organ or tissue) across the tenfold range of doses. Collectively, these data provide substantial novel evidence for the potential utility of CERE-120 as a novel treatment for PD and support ongoing clinical trials testing CERE-120 in PD patients.


Asunto(s)
Cuerpo Estriado/metabolismo , Dependovirus/genética , Vectores Genéticos , Neurturina/genética , Transgenes , Animales , Cuerpo Estriado/enzimología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Inmunohistoquímica , Macaca fascicularis , Masculino , Fosforilación , Tirosina 3-Monooxigenasa/metabolismo
18.
Stroke ; 39(4): 1254-61, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18309153

RESUMEN

BACKGROUND AND PURPOSE: Insulin-like growth factor I (IGF-1) is a pleiotropic growth factor that has been demonstrated to protect against acute ischemic brain injury. Whether IGF-1 improves long-term functional outcome after ischemic stroke is not known. The aim of this study is to examine whether IGF-1 overexpression through adeno-associated virus (AAV) -mediated gene transfer enhances neurovascular remodeling and improves functional outcome in a mouse model of focal cerebral ischemia. METHODS: Long-term cerebral IGF-1 overexpression was achieved with the AAV transduction system through stereotaxic injection. Control mice were injected with AAV-green fluorescent protein or saline. Three weeks after gene transfer, the mice underwent permanent distal middle cerebral artery occlusion. Histological and behavioral analyses were performed at day 21 after middle cerebral artery occlusion. RESULTS: IGF-1 gene transfer compared with control treatment significantly improved motor performance assessed by sensorimotor tests. The functional recovery was accompanied by reduced volume of cerebral infarction. Immunohistochemical analysis with endothelial cell marker CD31 revealed that IGF-1 gene transfer potently increased neovessel formation in the periinfarct and injection needle tract area compared with AAV-green fluorescent protein transduction. Increased vascular density was associated with increased local vascular perfusion. Additionally, AAV-IGF-1 treatment enhanced neurogenesis in the subventricular zone compared with AAV-green fluorescent protein treatment. CONCLUSIONS: These data demonstrate that IGF-1 overexpression promoted long-lasting functional recovery after cerebral infarction. The improved functional performance was paralleled by enhanced neovascularization and neurogenesis.


Asunto(s)
Terapia Genética/métodos , Infarto de la Arteria Cerebral Media/terapia , Factor I del Crecimiento Similar a la Insulina/genética , Neovascularización Fisiológica/fisiología , Animales , Atrofia , División Celular/fisiología , Circulación Cerebrovascular/fisiología , Dependovirus/genética , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/patología , Ratones , Ratones Endogámicos , Neuronas/citología , Recuperación de la Función/fisiología , Transducción Genética
19.
Lancet Neurol ; 7(5): 400-8, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18387850

RESUMEN

BACKGROUND: There is an urgent need for therapies that slow or reverse the progression of Parkinson's disease (PD). Neurotrophic factors can improve the function of degenerating neurons and protect against further neurodegeneration, and gene transfer might be a means to deliver effectively these factors to the brain. The aim of this study was to assess the safety, tolerability, and potential efficacy of gene delivery of the neurotrophic factor neurturin. METHODS: In this phase I, open-label clinical trial, 12 patients aged 35-75 years with a diagnosis of PD for at least 5 years in accordance with the UK Brain Bank Criteria received bilateral, stereotactic, intraputaminal injections of adeno-associated virus serotype 2-neurturin (CERE-120). The first six patients received doses of 1.3x10(11) vector genomes (vg)/patient, and the next six patients received 5.4x10(11) vg/patient. This trial is registered with ClinicalTrials.gov, number NCT00252850. FINDINGS: The procedure was well tolerated. Extensive safety monitoring in all patients revealed no clinically significant adverse events at 1 year. Several secondary measures of motor function showed improvement at 1 year; for example, a mean improvement in the off-medication motor subscore of the Unified Parkinson's Disease Rating Scale (UPDRS) of 14 points (SD 8; p=0.000121 [36% mean increase; p=0.000123]) and a mean increase of 2.3 h (2; 25% group mean increase; p=0.0250) in on time without troublesome dyskinesia were seen. Improvements in several secondary measures were not significant, including the timed walking test in the off condition (p=0.053), the Purdue pegboard test of hand dexterity (p=0.318), the reduction in off time (p=0.105), and the activities of daily living subscore (part II) of the UPDRS (p=0.080). (18)F-levodopa-uptake PET did not change after treatment with either dose of CERE-120. INTERPRETATION: The initial data support the safety, tolerability, and potential efficacy of CERE-120 as a possible treatment for PD; however, these results must be viewed as preliminary until data from blinded, controlled clinical trials are available. FUNDING: Ceregene; Michael J Fox Foundation for Parkinson's Research.


Asunto(s)
Dependovirus/fisiología , Terapia Genética/métodos , Neurturina/uso terapéutico , Enfermedad de Parkinson/terapia , Putamen/efectos de los fármacos , Adulto , Anciano , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos , Femenino , Vectores Genéticos/fisiología , Humanos , Masculino , Persona de Mediana Edad , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Neurturina/biosíntesis , Neurturina/genética , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología
20.
Mol Ther ; 15(1): 62-8, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17164776

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) or its naturally occurring analog, neurturin (NTN), can potentially improve the function and delay the rate of degeneration of dopaminergic neurons in Parkinson's disease (PD). However, their delivery to the central nervous system has proven to be a significant challenge. Viral vector-mediated gene transfer offers a practical means to continuously supply neurotrophic factors in targeted areas of the brain. CERE-120 is an adeno-associated viral vector encoding NTN, developed for the treatment of PD. We found that the kinetics and pattern of NTN expression in the rat striatum following injection of CERE-120 is rapid, increases significantly up to 4 weeks, and exhibits a stable volume of distribution thereafter for at least 1 year, the longest time-point evaluated. Quantitative enzyme-linked immunosorbent assay confirmed that steady-state levels are maintained from 4 weeks onward. We demonstrated that NTN volume of distribution can be controlled by varying the dose of vector injected and that NTN delivered via CERE-120 was bioactive, as evidenced by the neuroprotection of DA neurons in the rat 6-hydroxydopamine lesion model. These data provided the foundation for further non-clinical development of CERE-120, leading to an ongoing clinical trial in PD patients.


Asunto(s)
Dependovirus/genética , Dopamina/metabolismo , Vectores Genéticos/genética , Degeneración Nerviosa/genética , Neurturina/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/terapia , Animales , Línea Celular , Modelos Animales de Enfermedad , Dopamina/análogos & derivados , Expresión Génica , Terapia Genética , Genoma Viral/genética , Humanos , Cinética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Degeneración Nerviosa/terapia , Neurturina/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA