Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Brain Commun ; 2(2): fcaa046, 2020.
Article in English | MEDLINE | ID: mdl-33392501

ABSTRACT

There is an increasing need for improved endpoints to assess clinical trial effects in Parkinson's disease. We propose the Parkinson's Disease Comprehensive Response as a novel weighted composite endpoint integrating changes measured in three established Parkinson's outcomes, including: OFF state Movement Disorder Society Unified Parkinson's Disease Rating Scale Motor Examination scores; Motor Experiences of Daily Living scores; and total good-quality ON time per day. The data source for the initial development of the composite described herein was a recent Phase II trial of glial cell line-derived neurotrophic factor. A wide range of clinically derived relative weights was assessed to normalize for differentially scoring base rates with each endpoint component. The Parkinson's disease comprehensive response, in contrast to examining practically defined OFF state Unified Parkinson's Disease Rating Scale Motor Examination scores alone, showed stability over 40 weeks in placebo patients, and all 432 analyses in this permutation exercise yielded significant differences in favour of glial cell line-derived neurotrophic factor. The findings were consistent with results obtained employing three different global statistical test methodologies and with patterns of intra-patient change. Based on our detailed analyses, we conclude it worth prospectively evaluating the clinical utility, validity and regulatory feasibility of using clinically supported final Parkinson's disease comprehensive response formulas (for both the Unified Parkinson's Disease Rating Scale-based and Movement Disorders Society-Unified Parkinson's Disease Rating Scale-based versions) in future disease-modifying Parkinson's trials. Whilst the data source employed in the initial development of this weighted composite score is from a recent Phase II trial of glial cell line-derived neurotrophic factor, we wish to stress that the results are not described to provide post hoc evidence of the efficacy of glial cell line-derived neurotrophic factor but rather are presented to further the debate of how current regulatory approved rating scales may be combined to address some of the recognized limitations of using individual scales in isolation.

2.
J Parkinsons Dis ; 9(2): 301-313, 2019.
Article in English | MEDLINE | ID: mdl-30829619

ABSTRACT

BACKGROUND: Intraputamenal glial cell line-derived neurotrophic factor (GDNF), administered every 4 weeks to patients with moderately advanced Parkinson's disease, did not show significant clinical improvements against placebo at 40 weeks, although it significantly increased [18F]DOPA uptake throughout the entire putamen. OBJECTIVE: This open-label extension study explored the effects of continued (prior GDNF patients) or new (prior placebo patients) exposure to GDNF for another 40 weeks. METHODS: Using the infusion protocol of the parent study, all patients received GDNF without disclosing prior treatment allocations (GDNF or placebo). The primary outcome was the percentage change from baseline to Week 80 in the OFF state Unified Parkinson's Disease Rating Scale (UPDRS) motor score. RESULTS: All 41 parent study participants were enrolled. The primary outcome decreased by 26.7±20.7% in patients on GDNF for 80 weeks (GDNF/GDNF; N = 21) and 27.6±23.6% in patients on placebo for 40 weeks followed by GDNF for 40 weeks (placebo/GDNF, N = 20; least squares mean difference: 0.4%, 95% CI: -13.9, 14.6, p = 0.96). Secondary endpoints did not show significant differences between the groups at Week 80 either. Prespecified comparisons between GDNF/GDNF at Week 80 and placebo/GDNF at Week 40 showed significant differences for mean OFF state UPDRS motor (-9.6±6.7 vs. -3.8±4.2 points, p = 0.0108) and activities of daily living score (-6.9±5.5 vs. -1.0±3.7 points, p = 0.0003). No treatment-emergent safety concerns were identified. CONCLUSIONS: The aggregate study results, from the parent and open-label extension suggest that future testing with GDNF will likely require an 80- rather than a 40-week randomized treatment period and/or a higher dose.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Parkinson Disease/drug therapy , Putamen/diagnostic imaging , Antiparkinson Agents/therapeutic use , Dihydroxyphenylalanine/analogs & derivatives , Female , Fluorine Radioisotopes , Humans , Levodopa/therapeutic use , Male , Middle Aged , Parkinson Disease/diagnostic imaging , Parkinson Disease/metabolism , Positron-Emission Tomography , Putamen/metabolism , Randomized Controlled Trials as Topic
3.
Brain ; 142(3): 512-525, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30808022

ABSTRACT

We investigated the effects of glial cell line-derived neurotrophic factor (GDNF) in Parkinson's disease, using intermittent intraputamenal convection-enhanced delivery via a skull-mounted transcutaneous port as a novel administration paradigm to potentially afford putamen-wide therapeutic delivery. This was a single-centre, randomized, double-blind, placebo-controlled trial. Patients were 35-75 years old, had motor symptoms for 5 or more years, and presented with moderate disease severity in the OFF state [Hoehn and Yahr stage 2-3 and Unified Parkinson's Disease Rating Scale motor score (part III) (UPDRS-III) between 25 and 45] and motor fluctuations. Drug delivery devices were implanted and putamenal volume coverage was required to exceed a predefined threshold at a test infusion prior to randomization. Six pilot stage patients (randomization 2:1) and 35 primary stage patients (randomization 1:1) received bilateral intraputamenal infusions of GDNF (120 µg per putamen) or placebo every 4 weeks for 40 weeks. Efficacy analyses were based on the intention-to-treat principle and included all patients randomized. The primary outcome was the percentage change from baseline to Week 40 in the OFF state (UPDRS-III). The primary analysis was limited to primary stage patients, while further analyses included all patients from both study stages. The mean OFF state UPDRS motor score decreased by 17.3 ± 17.6% in the active group and 11.8 ± 15.8% in the placebo group (least squares mean difference: -4.9%, 95% CI: -16.9, 7.1, P = 0.41). Secondary endpoints did not show significant differences between the groups either. A post hoc analysis found nine (43%) patients in the active group but no placebo patients with a large clinically important motor improvement (≥10 points) in the OFF state (P = 0.0008). 18F-DOPA PET imaging demonstrated a significantly increased uptake throughout the putamen only in the active group, ranging from 25% (left anterior putamen; P = 0.0009) to 100% (both posterior putamina; P < 0.0001). GDNF appeared to be well tolerated and safe, and no drug-related serious adverse events were reported. The study did not meet its primary endpoint. 18F-DOPA imaging, however, suggested that intermittent convection-enhanced delivery of GDNF produced a putamen-wide tissue engagement effect, overcoming prior delivery limitations. Potential reasons for not proving clinical benefit at 40 weeks are discussed.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Adult , Aged , Double-Blind Method , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Infusion Pumps, Implantable , Male , Middle Aged , Neuroglia/metabolism , Placebo Effect , Treatment Outcome
4.
Arch Toxicol ; 92(7): 2353-2367, 2018 07.
Article in English | MEDLINE | ID: mdl-29785638

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) has demonstrated neurorestorative and neuroprotective effects in rodent and nonhuman primate models of Parkinson's disease. However, continuous intraputamenal infusion of GDNF (100 µg/day) resulted in multifocal cerebellar Purkinje cell loss in a 6-month toxicity study in rhesus monkeys. It was hypothesized that continuous leakage of GDNF into the cerebrospinal fluid compartment during the infusions led to down-regulation of GDNF receptors on Purkinje cells, and that subsequent acute withdrawal of GDNF then mediated the observed cerebellar lesions. Here we present the results of a 9-month toxicity study in which rhesus monkeys received intermittent intraputamenal infusions via convection-enhanced delivery. Animals were treated with GDNF (87.1 µg; N = 14) or vehicle (N = 6) once every 4 weeks for a total of 40 weeks (11 treatments). Four of the GDNF-treated animals were utilized in a satellite study assessing the impact of concomitant catheter repositioning prior to treatment. In the main study, eight animals (5 GDNF, 3 control) were euthanized at the end of the treatment period, along with the four satellite study animals, while the remaining eight animals (5 GDNF, 3 control) were euthanized at the end of a 12-week recovery period. There were no GDNF-related adverse effects and in particular, no GDNF-related microscopic findings in the brain, spinal cord, dorsal root ganglia, or trigeminal ganglia. Therefore, 87.1 µg/4 weeks is considered the no observed adverse effect level for GDNF in rhesus monkeys receiving intermittent, convection-enhanced delivery of GDNF for 9 months.


Subject(s)
Cerebellum/drug effects , Drug Delivery Systems/methods , Glial Cell Line-Derived Neurotrophic Factor/toxicity , Neuroprotective Agents/toxicity , Putamen/drug effects , Animals , Convection , Drug Administration Schedule , Drug Delivery Systems/instrumentation , Drug Evaluation, Preclinical , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Infusion Pumps, Implantable , Macaca mulatta , Male , Neuroprotective Agents/administration & dosage , No-Observed-Adverse-Effect Level , Toxicity Tests, Chronic
5.
Neurotoxicology ; 52: 46-56, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26535469

ABSTRACT

Recombinant-methionyl human glial cell line-derived neurotrophic factor (GDNF) is known for its neurorestorative and neuroprotective effects in rodent and primate models of Parkinson's disease (PD). When administered locally into the putamen of Parkinsonian subjects, early clinical studies showed its potential promise as a disease-modifying agent. However, the development of GDNF for the treatment of PD has been significantly clouded by findings of cerebellar toxicity after continuous intraputamenal high-dose administration in a 6-month treatment/3-month recovery toxicology study in rhesus monkeys. Specifically, multifocal cerebellar Purkinje cell loss affecting 1-21% of the cerebellar cortex was observed in 4 of 15 (26.7%; 95% confidence interval [CI]: 10.5-52.4%) animals treated at the highest dose level tested (3000µg/month). No cerebellar toxicity was observed at lower doses (450 and 900µg/month) in the same study, or at similar or higher doses (up to 10,000µg/month) in subchronic or chronic toxicology studies testing intermittent intracerebroventricular administration. While seemingly associated with the use of GDNF, the pathogenesis of the cerebellar lesions has not been fully understood to date. This review integrates available information to evaluate potential pathogenic mechanisms and provide a consolidated assessment of the findings. While other explanations are considered, the existing evidence is most consistent with the hypothesis that leakage of GDNF into cerebrospinal fluid during chronic infusions into the putamen down-regulates GDNF receptors on Purkinje cells, and that subsequent acute withdrawal of GDNF generates the observed lesions. The implications of these findings for clinical studies with GDNF are discussed.


Subject(s)
Cerebellum/drug effects , Glial Cell Line-Derived Neurotrophic Factor/adverse effects , Animals , Cerebellum/pathology , Cerebellum/physiopathology , Dose-Response Relationship, Drug , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Humans , Infusions, Intraventricular , Microinjections , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/adverse effects , Putamen/drug effects
6.
PLoS One ; 8(3): e56186, 2013.
Article in English | MEDLINE | ID: mdl-23526931

ABSTRACT

BACKGROUND: Despite promising early results, clinical trials involving the continuous delivery of recombinant methionyl human glial cell line-derived neurotrophic factor (r-metHuGDNF) into the putamen for the treatment of Parkinson's disease have shown evidence of poor distribution and toxicity due to point-source accumulation. Convection-enhanced delivery (CED) has the potential to facilitate more widespread and clinically effective drug distribution. AIMS: We investigated acute CED of r-metHuGDNF into the striatum of normal rats in order to assess tissue clearance, toxicity (neuron loss, gliosis, microglial activation, and decreases in synaptophysin), synaptogenesis and neurite-outgrowth. We investigated a range of clinically relevant infused concentrations (0.1, 0.2, 0.6 and 1.0 µg/µL) and time points (2 and 4 weeks) in order to rationalise a dosing regimen suitable for clinical translation. RESULTS: Two weeks after single dose CED, r-metHuGDNF was below the limit of detection by ELISA but detectable by immunohistochemistry when infused at low concentrations (0.1 and 0.2 µg/µL). At these concentrations, there was no associated neuronal loss (neuronal nuclei, NeuN, immunohistochemistry) or synaptic toxicity (synaptophysin ELISA). CED at an infused concentration of 0.2 µg/µL was associated with a significant increase in synaptogenesis (p<0.01). In contrast, high concentrations of r-metHuGDNF (above 0.6 µg/µL) were associated with neuronal and synaptic toxicity (p<0.01). Markers for gliosis (glial fibrillary acidic protein, GFAP) and microglia (ionized calcium-binding adapter molecule 1, Iba1) were restricted to the needle track and the presence of microglia had diminished by 4 weeks post-infusion. No change in neurite outgrowth (Growth associated protein 43, GAP43, mRNA) compared to artificial cerebral spinal fluid (aCSF) control was observed with any infused concentration. CONCLUSION: The results of this study suggest that acute CED of low concentrations of GDNF, with dosing intervals determined by tissue clearance, has most potential for effective clinical translation by optimising distribution and minimising the risk of toxic accumulation.


Subject(s)
Corpus Striatum/drug effects , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Animals , Cell Death/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Glial Cell Line-Derived Neurotrophic Factor/toxicity , Humans , Infusion Pumps , Male , Metabolic Clearance Rate , Neurogenesis/drug effects , Neuroglia/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Rats , Rats, Wistar , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/toxicity , Tissue Distribution
7.
J Neurosci Methods ; 196(1): 118-23, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21187114

ABSTRACT

Failures of recent convection-enhanced delivery (CED) trials have underscored the need for improved CED cannulae. This study presents a novel in-dwelling cannula targeted for use in upcoming neuro-oncological or neurodegeneration trials. The cannula is reflux-resistant and easily integrated into clinical workflows. The cannula was evaluated in non-human primate CED studies performed with magnetic resonance imaging (MRI) to validate the effectiveness and safety of the infusion.


Subject(s)
Catheterization/instrumentation , Drug Delivery Systems/instrumentation , Equipment Design , Animals , Brain/anatomy & histology , Contrast Media/administration & dosage , Convection , Follow-Up Studies , Gadolinium , Heterocyclic Compounds/administration & dosage , Macaca mulatta , Magnetic Resonance Imaging , Organometallic Compounds/administration & dosage , Reproducibility of Results
8.
Clin Neuropharmacol ; 27(6): 281-92, 2004.
Article in English | MEDLINE | ID: mdl-15613933

ABSTRACT

The neurobehavioral and pathologic features of Parkinson disease dementia (PDD) are virtually identical to those of dementia with Lewy bodies (DLB), suggesting that they represent different phenotypes of the same underlying disease. Both are characterized clinically by a "frontal-subcortical" dementia, fluctuating confusion, and, often, psychotic symptoms. Pathologically they are characterized by disseminated Lewy bodies and multiple transmitter deficits. These dementias with Lewy bodies constitute the second leading cause of dementia after Alzheimer disease (AD), and are thus an important treatment target. No drug has yet been approved for these indications, but treatment options are emerging. This paper addresses the conduct of clinical trials for this indication, including definition of target populations, screening metrics, outcome measures, and clinical trial designs. As the pathophysiology of these cognitive and behavioral changes becomes better understood, symptomatic as well as disease-modifying therapy may become possible, requiring an inclusive and consistent approach to clinical trials in this area.


Subject(s)
Clinical Trials as Topic/methods , Lewy Body Disease/physiopathology , Cognition/physiology , Humans , Lewy Body Disease/epidemiology , Lewy Body Disease/therapy , Motor Activity/physiology , Neuropsychological Tests , Parkinson Disease/physiopathology , Quality of Life , Research Design
SELECTION OF CITATIONS
SEARCH DETAIL
...