Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 147: 28-36, 2019 03 15.
Article in English | MEDLINE | ID: mdl-29857941

ABSTRACT

Parkinson's disease (PD) is a disorder affecting dopamine neurons for which there is no cure. Glial cell line-derived neurotrophic factor (GDNF) and the closely related protein neurturin are two trophic factors with demonstrated neuroprotective and neurorestorative properties on dopamine neurons in multiple animal species. However, GDNF and neurturin Phase-2 clinical trials have failed to demonstrate a significant level of improvement over placebo controls. Insufficient drug distribution in the brain parenchyma has been proposed as a major contributing factor for the lack of clinical efficacy in the Phase-2 trial patients. To address this issue, a novel mammalian cell-derived variant form of GDNF (GDNFv) was designed to promote better tissue distribution by reducing its heparin binding to the extracellular matrix and key amino acids were substituted to enhance its chemical stability. Administration of this fully glycosylated GDNFv in the normal rat striatum increased dopamine turnover and produced significantly greater brain distribution than E. coli-produced wildtype GDNF (GDNFwt). Intrastriatal GDNFv also protected midbrain dopamine neuron function in 6-hydroxydopamine-lesioned rats. Studies conducted in normal adult rhesus macaques support that GDNFv was well tolerated in all animals and demonstrated a greater volume of distribution than GDNFwt in the brain following intrastriatal infusion. Importantly, favorable physiological activity of potential therapeutic value was maintained in this variant trophic factor with significant target activation in GDNFv recipients as indicated by dopamine turnover modulation. These data suggest that GDNFv may be a promising drug candidate for the treatment of PD. Additional studies are needed in non-human primates with dopamine depletion. This article is part of the Special Issue entitled 'Drug Repurposing: old molecules, new ways to fast track drug discovery and development for CNS disorders'.


Subject(s)
Brain/metabolism , Dopamine/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Neurturin/pharmacology , Animals , Brain/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Humans , Macaca mulatta , Neurturin/pharmacokinetics , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Rats , Rats, Sprague-Dawley , Tissue Distribution
2.
Mol Vis ; 24: 733-745, 2018.
Article in English | MEDLINE | ID: mdl-30581280

ABSTRACT

Purpose: To evaluate the potential of a poly(lactic-co-glycolic acid) (PLGA)-based slow release formulation of glial cell line-derived neurotrophic factor (GDNF) alone or in combination with melatonin to rescue photoreceptors in a mouse model of retinal degeneration. Methods: GDNF and GDNF/melatonin-loaded PLGA microspheres (MSs) were prepared using a solid-in-oil-in-water emulsion solvent extraction-evaporation technique. A combination of PLGA and vitamin E (VitE) was used to create the microcarriers. The structure, particle size, encapsulation efficiency, and in vitro release profile of the microparticulate formulations were characterized. Microparticulate systems (non-loaded, GDNF, and GDNF/melatonin-loaded MSs) were administered intravitreally to 3-week-old rhodopsin knockout mice (rho (-/-); n=7). The functional neuroprotective effect was assessed with electroretinography at 6, 9, and 12 weeks old. The rescue of the structure was determined with photoreceptor quantification at 12 weeks (9 weeks after administration of MSs). Immunohistochemistry for photoreceptor, glial, and proliferative markers was also performed. Results: The microspheres were able to deliver GDNF or to codeliver GDNF and melatonin in a sustained manner. Intravitreal injection of GDNF or GDNF/melatonin-loaded MSs led to partial functional and structural rescue of photoreceptors compared to blank microspheres or vehicle. No significant intraocular inflammatory reaction was observed after intravitreal injection of the microspheres. Conclusions: A single intravitreal injection of GDNF or GDNF/melatonin-loaded microspheres in the PLGA/VitE combination promoted the rescue of the photoreceptors in rho (-/-) mice. These intraocular drug delivery systems enable the efficient codelivery of therapeutically active substances for the treatment of retinal diseases.


Subject(s)
Delayed-Action Preparations/pharmacokinetics , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Melatonin/pharmacokinetics , Retina/drug effects , Retinal Degeneration/therapy , Rhodopsin/genetics , Animals , Delayed-Action Preparations/chemistry , Disease Models, Animal , Drug Combinations , Drug Compounding/methods , Drug Liberation , Electroretinography , Gene Expression , Intravitreal Injections , Mice , Mice, Knockout , Microspheres , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Retina/metabolism , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Rhodopsin/agonists , Rhodopsin/deficiency , Vitamin E/chemistry , Vitreous Body
3.
Acta Biomater ; 69: 146-155, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29330037

ABSTRACT

Facial nerve injury caused by traffic accidents or operations may reduce the quality of life in patients, and recovery following the injury presents unique clinical challenges. Glial cell-derived neurotrophic factor (GDNF) is important in nerve regeneration; however, soluble GDNF rapidly diffuses into body fluids, making it difficult to achieve therapeutic efficacy. In this work, we developed a rat tail derived collagen conduit to connect nerve defects in a simple and safe manner. GDNF was immobilized in the collagen conduits via chemical conjugation to enable controlled release of GDNF. The GDNF delivery system prevented rapid diffusion from the site without impacting bioactivity of GDNF; degradation of the collagen conduit was inhibited owing to the chemical conjugation. The artificial nerve conduit was then used to examine facial nerve regeneration across a facial nerve defect. Following transplantation, the artificial nerve conduits degraded gradually without causing dislocations and serious inflammation, with good integration into the host tissue. Functional and histological tests indicated that the artificial nerve conduits were able to guide the axons to grow through the defect, reaching the distal stumps. The degree of nerve regeneration in the group that was treated with the artificial nerve conduit approached that of the autograft group, and exceeded that of the other conduit grafted groups. STATEMENT OF SIGNIFICANCE: In this study, we developed artificial nerve conduits consisting of GDNF immobilized on collagen, with the aim of providing an environment for nerve regeneration. Our results show that the artificial nerve conduits guided the regeneration of axons to the distal nerve segment. GDNF was immobilized stably in the artificial nerve conduits, and therefore retained a sufficient concentration at the target site to effectively promote the regeneration process. The artificial nerve conduits exhibited good biocompatibility and facilitated nerve regeneration and functional recovery with an efficacy that was close to that of an autograft, and better than that of the other conduit grafted groups. Our approach provides an effective delivery system that overcomes the rapid diffusion of GDNF in body fluids, promoting peripheral nerve regeneration. The artificial nerve conduit therefore qualifies as a putative candidate material for the fabrication of peripheral nerve reconstruction devices.


Subject(s)
Absorbable Implants , Collagen , Facial Nerve Injuries/therapy , Facial Nerve/physiology , Glial Cell Line-Derived Neurotrophic Factor , Nerve Regeneration/drug effects , Animals , Collagen/chemistry , Collagen/pharmacokinetics , Collagen/pharmacology , Drug Implants/chemistry , Drug Implants/pharmacokinetics , Drug Implants/pharmacology , Facial Nerve Injuries/metabolism , Facial Nerve Injuries/pathology , Female , Glial Cell Line-Derived Neurotrophic Factor/chemistry , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Rats , Rats, Sprague-Dawley
4.
Med Biol Eng Comput ; 55(12): 2069-2077, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28493093

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) is a potential therapy for Parkinson's disease (PD) promoting survival and functional recovery of dopaminergic neurons when delivered to the degenerated striatum. To study the aspects of intraputamenal delivery of GDNF, a mathematical model of recombinant methionyl human GDNF (r-metHuGDNF) convection in the human putamen has been developed. The convection-enhanced delivery infusions of r-metHuGDNF were simulated at rates up to 5 µL/min. The high-rate infusions (≥1 µL/min) permit rapid and uniform distribution of drug with up to 75% of the distribution volume having a concentration within 5% of the infusate concentration. No relevant differences in distribution at infusion rates of 3 and 5 µL/min were found. The patterns of GDNF distribution were analyzed in relation to the anatomy of the posterior dorsal putamen, and a cylindrical shape was found to be preferable considering risks of target overflow. A magnetic resonance (MR) tracer Gd-DTPA (Magnevist®) was evaluated as a surrogate in clinical studies, and the most accurate prediction of GDNF distribution was calculated immediately after infusion. The clearance of GDNF from the striatum is confirmed to be slow, with a half-life of ca. 19 h.


Subject(s)
Computer Simulation , Glial Cell Line-Derived Neurotrophic Factor , Models, Biological , Putamen , Corpus Striatum/diagnostic imaging , Corpus Striatum/metabolism , Gadolinium DTPA/administration & dosage , Gadolinium DTPA/pharmacokinetics , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Humans , Putamen/diagnostic imaging , Putamen/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics
5.
Eur J Pharm Sci ; 103: 19-26, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28259830

ABSTRACT

Local long-term delivery of glial cell line derived neurotrophic factor (GDNF) from vitamin E/poly-lactic-co-glycolic acid microspheres (MSs) protects retinal ganglion cells in an animal model of glaucoma for up to 11weeks. However, the pharmacokinetics of GDNF after intravitreal injection of MSs is not known. We evaluated the GDNF levels after a single intravitreal injection of GDNF/VitE MSs. Biodegradable MSs were prepared by the solid-oil-in-water emulsion-solvent evaporation technique and characterized. Rabbits received a single intravitreal injection (50µL) of GDNF/VitE MSs (4%w/v; 24 right eyes; 74.85ng GDNF), blank MSs (4%w/v; 24 left eyes), and balanced salt solution (4 eyes). Two controls eyes received no injections. At 24h, 1, 4, 6, 8, 12, 18, and 24weeks after injection, the eyes were enucleated, and the intravitreal GDNF levels were quantified. Pharmacokinetic data were analysed according to non-compartmental model. Intraocular GDNF levels of 717.1±145.1pg/mL were observed at 24h for GDNF-loaded MSs, followed by a plateau (745.3±25.5pg/mL) until day 28. After that, a second plateau (17.4±3.7pg/mL) occurred from 8 to 24weeks post-injection, significantly higher than the basal levels. Eyes injected with GDNF/vitE and Blank-MSs did not show any abnormalities during the six-months follow up after administration. The single injection of GDNF/VitE MSs provided a sustained controlled release of the neurotrophic factor in a controlled fashion for up to six months.


Subject(s)
Glaucoma/drug therapy , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Animals , Drug Carriers , Drug Liberation , Female , Glial Cell Line-Derived Neurotrophic Factor/chemistry , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Humans , Intravitreal Injections , Lactic Acid , Microspheres , Particle Size , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Rabbits , Vitamin E
6.
Sci Rep ; 6: 32292, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27558932

ABSTRACT

Recurrent laryngeal nerve (RLN) injury remains a challenge due to the lack of effective treatments. In this study, we established a new drug delivery system consisting of a tube of Heal-All Oral Cavity Repair Membrane loaded with laminin and neurotrophic factors and tested its ability to promote functional recovery following RLN injury. We created recombinant fusion proteins consisting of brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) fused to laminin-binding domains (LBDs) in order to prevent neurotrophin diffusion. LBD-BDNF, LBD-GDNF, and laminin were injected into a collagen tube that was fitted to the ends of the transected RLN in rats. Functional recovery was assessed 4, 8, and 12 weeks after injury. Although vocal fold movement was not restored until 12 weeks after injury, animals treated with the collagen tube loaded with laminin, LBD-BDNF and LBD-GDNF showed improved recovery in vocalisation, arytenoid cartilage angles, compound muscle action potentials and regenerated fibre area compared to animals treated by autologous nerve grafting (p < 0.05). These results demonstrate the drug delivery system induced nerve regeneration following RLN transection that was superior to that induced by autologus nerve grafting. It may have potential applications in nerve regeneration of RLN transection injury.


Subject(s)
Brain-Derived Neurotrophic Factor , Collagen , Glial Cell Line-Derived Neurotrophic Factor , Laminin , Laryngeal Nerves/physiology , Lingual Nerve Injuries/therapy , Nerve Regeneration/drug effects , Tissue Scaffolds/chemistry , Animals , Brain-Derived Neurotrophic Factor/chemistry , Brain-Derived Neurotrophic Factor/pharmacokinetics , Brain-Derived Neurotrophic Factor/pharmacology , Collagen/chemistry , Collagen/pharmacokinetics , Collagen/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/chemistry , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Laminin/chemistry , Laminin/pharmacokinetics , Laminin/pharmacology , Lingual Nerve Injuries/metabolism , Lingual Nerve Injuries/pathology , Male , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology
7.
Mol Neurobiol ; 52(2): 846-58, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26041662

ABSTRACT

Administration of various neurotrophic factors is a promising strategy against Parkinson's disease (PD). An intrastriatal infusion of 6-hydroxidopamine (6-OHDA) in rats is a suitable model to study PD. This work aims to describe stereological parameters regarding rostro-caudal gradient, in order to characterize the model and verify its suitability for elucidating the benefits of therapeutic strategies. Administration of 6-OHDA induced a reduction in tyrosine hidroxylase (TH) reactivity in the dorsolateral part of the striatum, being higher in the caudal section than in the rostral one. Loss of TH-positive neurons and axodendritic network was highly significant in the external third of substantia nigra (e-SN) in the 6-OHDA group versus the saline one. After the administration of nanospheres loaded with neurotrophic factors (NTF: vascular endothelial growth factor (VEGF) + glial cell line-derived neurotrophic factor (GDNF)), parkinsonized rats showed more TH-positive fibers than those of control groups; this recovery taking place chiefly in the rostral sections. Neuronal density and axodendritic network in e-SN was more significant than in the entire SN; the topographical analysis showed that the highest difference between NTF versus control group was attained in the middle section. A high number of bromodeoxyuridine (BrdU)-positive cells were found in sub- and periventricular areas in the group receiving NTF, where most of them co-expressed doublecortin. Measurements on the e-SN achieved more specific and significant results than in the entire SN. This difference in rostro-caudal gradients underpins the usefulness of a topological approach to the assessment of the lesion and therapeutic strategies. Findings confirmed the neurorestorative, neurogenic, and synergistic effects of VEGF+GDNF administration.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Parkinsonian Disorders/drug therapy , Vascular Endothelial Growth Factor A/therapeutic use , Animals , Corpus Striatum/drug effects , Corpus Striatum/enzymology , DNA Replication , Doublecortin Domain Proteins , Doublecortin Protein , Drug Synergism , Drug Therapy, Combination , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Glial Fibrillary Acidic Protein/analysis , Injections, Intralesional , Male , Microtubule-Associated Proteins/analysis , Nanocapsules , Nerve Tissue Proteins/analysis , Neuroglia/chemistry , Neuroglia/drug effects , Neuroglia/ultrastructure , Neurons/chemistry , Neurons/drug effects , Neurons/ultrastructure , Neuropeptides/analysis , Oxidopamine/toxicity , Parkinsonian Disorders/pathology , Rats , Rats, Sprague-Dawley , Tissue Distribution , Tyrosine 3-Monooxygenase/analysis , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/pharmacokinetics
8.
Adv Healthc Mater ; 3(5): 761-74, 2014 May.
Article in English | MEDLINE | ID: mdl-24596339

ABSTRACT

Brain repair following disease and injury is very limited due to difficulties in recruiting and mobilizing stem cells towards the lesion. More importantly, there is a lack of structural and trophic support to maintain viability of the limited stem/progenitor cells present. This study investigates the effectiveness of an injectable gelatin-based hydrogel in attracting neural progenitor cells (NPCs) from the subventricular zone (SVZ) towards the implant. Glial cell-line-derived neurotrophic factor (GDNF) encapsulated within the hydrogel and porosity within the hydrogel prevents glial scar formation. By directly targeting the hydrogel implant towards the SVZ, neuroblasts can actively migrate towards and along the implant tract. Significantly more doublecortin (DCX)-positive neuroblasts surround implants at 7 d post-implantation (dpi) compared with lesion alone controls, an effect that is enhanced when GDNF is incorporated into the hydrogels. Neuroblasts are not observed at the implant boundary at 21 dpi, indicating that neuroblast migration has halted, and neuroblasts have either matured or have not survived. The development of an injectable gelatin-based hydrogel has significant implications for the treatment of some neurodegenerative diseases and brain injuries. The ability of GDNF and porosity to effectively prevent glial scar formation will allow better integration and interaction between the implant and surrounding neural tissue.


Subject(s)
Cell Movement/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Neural Stem Cells/drug effects , Animals , Astrocytes/cytology , Astrocytes/drug effects , Doublecortin Protein , Gelatin/administration & dosage , Gelatin/chemistry , Gelatin/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/chemistry , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Hydrogels/administration & dosage , Hydrogels/chemistry , Hydrogels/pharmacology , Male , Neural Stem Cells/cytology , Neurites/drug effects , Rats , Rats, Wistar
9.
Int J Mol Sci ; 15(3): 3612-23, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24583850

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) was encapsulated into liposomes in order to protect it from enzyme degradation in vivo and promote its permeability across the blood-brain barrier (BBB). In this study, GDNF conventional liposomes (GDNF-L) and GDNF target sterically stabilized liposomes (GDNF-SSL-T) were prepared. The average size of liposomes was below 90 nm. A primary model of BBB was established and evaluated by transendothelial electrical resistance (TEER) and permeability. This BBB model was employed to study the permeability of GDNF liposomes in vitro. The results indicated that the liposomes could enhance transport of GDNF across the BBB and GDNF-SSL-T had achieved the best transport efficacy. The distribution of GDNF liposomes was studied in vivo. Free GDNF and GDNF-L were eliminated rapidly in the circulation. GDNF-SSL-T has a prolonged circulation time in the blood and favorable brain delivery. The values of the area under the curve (AUC(0-1 h)) in the brain of GDNF-SSL-T was 8.1 times and 6.8 times more than that of free GDNF and GDNF-L, respectively. These results showed that GDNF-SSL-T realized the aim of targeted delivery of therapeutic proteins to central nervous system.


Subject(s)
Blood-Brain Barrier/metabolism , Capillary Permeability , Cell Membrane Permeability , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Animals , Astrocytes/metabolism , Biological Transport , Brain/blood supply , Capillaries/cytology , Cells, Cultured , Drug Delivery Systems/methods , Endothelial Cells/metabolism , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Liposomes , Models, Biological , Rats, Sprague-Dawley , Time Factors
10.
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
11.
Neuroimage ; 54 Suppl 1: S189-95, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20080195

ABSTRACT

Recently, we developed an MRI-based method that enables tracking of parenchymal infusions of therapeutic agents by inclusion of a contrast reagent in the infusate. We show that both liposomal Gadoteridol (GDL) and free Gadoteridol (Gd) can be used for MRI-monitored infusions into the non-human primate (NHP) putamen to predict the distribution of GDNF protein after convection-enhanced delivery (CED). GDNF and both MRI tracers showed good co-distribution within the putamen and other brain regions. Although the CED infusion technique can distribute GDNF protein over large brain regions, continuous administration of GDNF could cause undesired effects that could counteract the benefits of CED as demonstrated in this study when large volumes of GDNF were delivered that lead to GDNF leakage into CSF. These limitations can be addressed by employing an intermittent CED schedule that permits consistent target coverage without GDNF leakage into CSF or white matter. We present an approach intracranial GDNF infusions that can be optimized by means of real-time monitoring via MRI. Adoption of this new standard, along with advanced, reflux-resistant cannulae, may permit reconsideration of direct GDNF infusion into parenchyma as a clinical strategy, since previous clinical studies involving chronic infusion of recombinant glial cell line-derived neurotrophic factor (GDNF) to the putamen for the treatment of Parkinson's disease have yielded mixed results, a state of affairs that may in part be attributed to suboptimal infusion parameters.


Subject(s)
Contrast Media/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Heterocyclic Compounds/administration & dosage , Organometallic Compounds/administration & dosage , Putamen/metabolism , Surgery, Computer-Assisted/methods , Animals , Contrast Media/pharmacokinetics , Convection , Drug Delivery Systems/methods , Gadolinium , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Heterocyclic Compounds/pharmacokinetics , Macaca fascicularis , Magnetic Resonance Imaging , Male , Organometallic Compounds/pharmacokinetics , Tissue Distribution
12.
Exp Eye Res ; 91(6): 890-5, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20883692

ABSTRACT

The purpose of this study was to establish the intravitreal (ITV) pharmacokinetics of glial cell line-derived neurotrophic factor (GDNF) and observe possible complications after ITV injection. Twenty Danish landrace pigs and 34 eyes were included in the study; 30 were injected with 100 ng of GDNF, two controls were injected without GDNF, and two received no injection. At post-injection time points of 1, 2, 3, 6 hours (h), 1, 2, 4 or 7 days (d) eyes were enucleated and the ITV concentration of GDNF (cGDNF) was determined by enzyme-linked immunosorbent assay, and activity was tested using a retinal ganglion cell line (RGC5) bioassay. Indirect ophthalmoscopy, intraocular pressure assessment, and fundus photography were performed before enucleation. There was initial variability in the cGDNF, but after 24h GDNF was cleared in a monoexponential fashion with a half-life of 37 h (CL 33-43 h). Therapeutic concentrations were present for 15 d (CL 13-18d) when an extrapolation was done. GDNF-injected vitreous samples stimulated increased survival of RGC5s at 24h post-delivery (p=0.002) compared with no-GDNF vitreous controls. This effect was independent of intraocular incubation time when cGDNF was normalized to 5 ng/ml. A semi-logarithmic dose-response curve showed linearity between 0.1 and 10 ng/ml. None of the eyes showed any signs of inflammation or other complications. A single ITV GDNF injection of 100 ng leads to therapeutic levels for 15 days in the porcine eye. The GDNF was stable in the intraocular environment and no adverse events were observed. GDNF might therefore play a role in the future treatment of acute retinal damage.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Vitreous Body/metabolism , Animals , Cell Survival , Enzyme-Linked Immunosorbent Assay , Female , Half-Life , Intraocular Pressure , Intravitreal Injections , Ophthalmoscopy , Recombinant Proteins/pharmacokinetics , Retinal Ganglion Cells/cytology , Swine
14.
Neuropharmacology ; 58(7): 1114-21, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20153340

ABSTRACT

Convection-enhanced delivery (CED) of GDNF and NTN was employed to determine the tissue clearance of these factors from the rat striatum and the response of the dopaminergic system to a single infusion. Two doses of GDNF (15 and 3 microg) and NTN (10 microg and 2 microg) were infused into the rat striatum. Animals were euthanized 3, 7, 14, 21, and 28 days post-infusion. Brains were processed for ELISA, HPLC, and immunohistochemistry (IHC). Both doses of the infused GDNF resulted in a sharp increase in striatal GDNF levels followed by a rapid decrease between day 3 and 7. Interestingly, IHC revealed GDNF in the septum and the base of the brain 14 days after GDNF administration. Dopamine (DA) turnover was significantly increased in a dose-dependent manner for more than 7 days after a single GDNF infusion. NTN persisted in the brain for at least two weeks longer than GDNF. It also had more persistent effects on DA turnover, probably due to its precipitation in the brain at neutral pH after infusion. Our data suggest that daily or continuous dosing may not be necessary for delivering growth factors into the CNS.


Subject(s)
Brain/metabolism , Dopamine/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Neurturin/pharmacology , Neurturin/pharmacokinetics , Animals , Brain/drug effects , Chromatography, High Pressure Liquid , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Immunohistochemistry , Male , Neurturin/administration & dosage , Neurturin/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
15.
Drug Metab Dispos ; 38(4): 566-72, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20075191

ABSTRACT

Glial-derived neurotrophic factor (GDNF) is a potent neuroprotective agent for multiple brain disorders, including Parkinson's disease. However, GDNF drug development is difficult because GDNF does not cross the blood-brain barrier (BBB). To enable future drug development of GDNF in mouse models, the neurotrophin was re-engineered as an IgG fusion protein to enable penetration through the BBB after intravenous administration. The 134-amino acid GDNF was fused to the heavy chain of a chimeric monoclonal antibody (MAb) against the mouse transferrin receptor (TfR) designated the cTfRMAb. This antibody undergoes receptor-mediated transport across the BBB and acts as a molecular Trojan horse to ferry the GDNF into mouse brain. The cTfRMAb-GDNF fusion protein was expressed by stably transfected Chinese hamster ovary cells, affinity-purified, and the biochemical identity was confirmed by mouse IgG and GDNF Western blotting. The cTfRMAb-GDNF fusion protein was bifunctional and bound with high affinity to both the GDNF receptor alpha1, ED(50) = 1.7 +/- 0.2 nM, and the mouse TfR, ED(50) = 3.2 +/- 0.3 nM. The cTfRMAb-GDNF fusion protein was rapidly taken up by brain, and the brain uptake was 3.1 +/- 0.2% injected dose/g brain at 60 min after intravenous injection of a 1-mg/kg dose of the fusion protein. Brain capillary depletion analysis showed the majority of the fusion protein was transcytosed across the BBB with penetration into brain parenchyma. The brain uptake results indicate it is possible to achieve therapeutic elevations of GDNF in mouse brain with intravenous administration of the cTfRMAb-GDNF fusion protein.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Blood-Brain Barrier/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Animals , Blotting, Western , CHO Cells , Capillaries/metabolism , Cell Line , Cricetinae , Cricetulus , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Mice, Inbred C57BL , Neurons/metabolism , Radioligand Assay , Receptors, Transferrin/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Tetrahydrofolate Dehydrogenase/metabolism
16.
Biochem Biophys Res Commun ; 390(3): 947-51, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19852934

ABSTRACT

With a view toward improving delivery of exogenous glial cell line-derived neurotrophic factor (GDNF) to CNS motor neurons in vivo, we evaluated the bioavailability and pharmacological activity of a recombinant GDNF:tetanus toxin C-fragment fusion protein in mouse CNS. Following intramuscular injection, GDNF:TTC but not recombinant GDNF (rGDNF) produced strong GDNF immunostaining within ventral horn cells of the spinal cord. Intrathecal infusion of GDNF:TTC resulted in tissue concentrations of GDNF in lumbar spinal cord that were at least 150-fold higher than those in mice treated with rGDNF. While levels of immunoreactive choline acetyltransferase and GFRalpha-1 in lumbar cord were not altered significantly by intrathecal infusion of rGNDF, GDNF:TTC, or TTC, only rGDNF and GDNF:TTC caused significant weight loss following intracerebroventricular infusion. These studies indicate that insect cell-derived GDNF:TTC retains its bi-functional activity in mammalian CNS in vivo and improves delivery of GDNF to spinal cord following intramuscular- or intrathecal administration.


Subject(s)
Drug Delivery Systems , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Motor Neurons/metabolism , Peptide Fragments/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Spinal Cord/metabolism , Tetanus Toxin/administration & dosage , Animals , Biological Availability , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Peptide Fragments/pharmacokinetics , Recombinant Fusion Proteins/pharmacokinetics , Tetanus Toxin/pharmacokinetics
17.
Drug Metab Dispos ; 37(12): 2299-304, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19741036

ABSTRACT

The brain drug development of glial-derived neurotrophic factor (GDNF) is prevented by the lack of transport of this protein across the blood-brain barrier (BBB). GDNF transport across the BBB can be made possible by re-engineering the neurotrophin as a fusion protein with a genetically engineered monoclonal antibody (MAb) against the human insulin receptor (HIR), which crosses the BBB on the endogenous insulin receptor. The present work was designed to compare the BBB transport in vivo of GDNF and the HIR MAb-GDNF fusion protein. Owing to species specificity of HIR MAb binding to the insulin receptor, the present studies were performed in the adult rhesus monkey. The brain uptake of human IgG1 was determined to assess the uptake of a brain plasma volume marker. The brain clearance of GDNF was no different from the clearance of the IgG1, which indicated GDNF does not cross the primate BBB in vivo. In contrast, BBB transport of the HIR MAb-GDNF fusion protein was shown with film and emulsion autoradiography, as well as the capillary depletion method. In parallel with the increased brain uptake, fusion of the GDNF to the HIR MAb resulted in a decrease in the uptake of GDNF by liver, spleen, and kidney. Administration of the HIR MAb-GDNF fusion protein had no effect on glycemic control. The brain uptake parameters show that a systemic dose of the HIR MAb-GDNF fusion protein of 0.2 mg/kg may generate a 10-fold increase in the cerebral concentration of GDNF in the human brain.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Blood-Brain Barrier/metabolism , Capillary Permeability , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Immunoglobulin G/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Autoradiography , Biological Transport , Enzyme-Linked Immunosorbent Assay , Female , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Humans , Immunoglobulin G/administration & dosage , Injections, Intravenous , Iodine Radioisotopes , Macaca mulatta , Receptor, Insulin/immunology , Receptor, Insulin/metabolism , Recombinant Fusion Proteins/pharmacokinetics
18.
J Neurosci ; 29(30): 9651-9, 2009 Jul 29.
Article in English | MEDLINE | ID: mdl-19641128

ABSTRACT

Neurotrophic factors are promising candidates for the treatment of Parkinson's disease (PD). Mesencephalic astrocyte-derived neurotrophic factor (MANF) belongs to a novel evolutionarily conserved family of neurotrophic factors. We examined whether MANF has neuroprotective and neurorestorative effect in an experimental model of PD in rats. We also studied the distribution and transportation of intrastriatally injected MANF in the brain and compared it with glial cell line-derived neurotrophic factor (GDNF). Unilateral lesion of nigrostriatal dopaminergic system was induced by intrastriatal injection of 6-hydroxydopamine (6-OHDA). Amphetamine-induced turning behavior was monitored up to 12 weeks after the unilateral lesion. The local diffusion at the injection site and transportation profiles of intrastriatally injected MANF and GDNF were studied by immunohistochemical detection of the unlabeled growth factors as well as by autoradiographic and gamma counting detection of (125)I-labeled trophic factors. Intrastriatally injected MANF protected nigrostriatal dopaminergic nerves from 6-OHDA-induced degeneration as evaluated by counting tyrosine hydroxylase (TH)-positive cell bodies in the substantia nigra (SN) and TH-positive fibers in the striatum. More importantly, MANF also restored the function of the nigrostriatal dopaminergic system when administered either 6 h before or 4 weeks after 6-OHDA administration in the striatum. MANF was distributed throughout the striatum more readily than GDNF. The mechanism of MANF action differs from that of GDNF because intrastriatally injected (125)I-MANF was transported to the frontal cortex, whereas (125)I-GDNF was transported to the SN. Our results suggest that MANF is readily distributed throughout the striatum and has significant therapeutic potential for the treatment of PD.


Subject(s)
Nerve Tissue Proteins/administration & dosage , Neuroprotective Agents/administration & dosage , Parkinson Disease, Secondary/drug therapy , Animals , Corpus Striatum/drug effects , Corpus Striatum/physiopathology , Disease Models, Animal , Dopamine/metabolism , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Humans , Male , Motor Activity/drug effects , Nerve Degeneration/drug therapy , Nerve Growth Factors , Nerve Tissue Proteins/pharmacokinetics , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/pharmacokinetics , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Rats , Rats, Wistar , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Substantia Nigra/drug effects , Substantia Nigra/physiopathology , Time Factors
19.
Pharm Res ; 26(10): 2227-36, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19609743

ABSTRACT

PURPOSE: Glial-derived neurotrophic factor (GDNF) is a potential therapy for stroke, Parkinson's disease, or drug addiction. However, GDNF does not cross the blood-brain barrier (BBB). GDNF is re-engineered as a fusion protein with a chimeric monoclonal antibody (MAb) to the human insulin receptor (HIR), which acts as a molecular Trojan horse to deliver the GDNF across the BBB. The pharmacokinetics (PK), toxicology, and safety pharmacology of the HIRMAb-GDNF fusion protein were investigated in Rhesus monkeys. METHODS: The fusion protein was administered as an intravenous injection at doses up to 50 mg/kg over a 60 h period to 56 Rhesus monkeys. The plasma concentration of the HIRMAb-GDNF fusion protein was measured with a 2-site sandwich ELISA. RESULTS: No adverse events were observed in a 2-week terminal toxicology study, and no neuropathologic changes were observed. The PK analysis showed a linear relationship between plasma AUC and dose, a large systemic volume of distribution, as well as high clearance rates of 8-10 mL/kg/min. CONCLUSIONS: A no-observable-adverse-effect level is established in the Rhesus monkey for the acute administration of the HIRMAb-GDNF fusion protein. The fusion protein targeting the insulin receptor has a PK profile similar to a classical small molecule.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Blood-Brain Barrier/metabolism , Drug Delivery Systems/methods , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Recombinant Fusion Proteins/pharmacokinetics , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Blood-Brain Barrier/drug effects , Dose-Response Relationship, Drug , Female , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/adverse effects , Macaca mulatta , Male , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects
20.
Biotechnol Bioeng ; 100(2): 387-96, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18080333

ABSTRACT

Glial-derived neurotrophic factor (GDNF) is a neurotrophin that could be developed as a neurotherapeutic for Parkinson's disease, stroke, and motor neuron disease. However, GDNF does not cross the blood-brain barrier (BBB). Human GDNF was re-engineered by fusion of the mature GDNF protein to the carboxyl terminus of the chimeric monoclonal antibody (MAb) to the human insulin receptor (HIR). The HIRMAb-GDNF fusion protein is bi-functional, and both binds the HIR, to trigger receptor-mediated transport across the BBB, and binds the GDNF receptor (GFR)-alpha1, to activate GDNF neuroprotection pathways behind the BBB. COS cells were dual transfected with the heavy chain (HC) and light chain fusion protein expression plasmids, and the HC of the fusion protein was immunoreactive with antibodies to both human IgG and GDNF. The HIRMAb-GDNF fusion protein bound with high affinity to the extracellular domain of both the HIR, ED(50) = 0.87 +/- 0.13 nM, and the GFRalpha1, ED(50) = 1.68 +/- 0.17 nM. The HIRMAb-GDNF fusion protein activated luciferase gene expression in human neural SK-N-MC cells dual transfected with the c-ret kinase and a luciferase reporter gene under the influence of the rat tyrosine hydroxylase promoter, and the ED(50), 1.68 +/- 0.45 nM, was identical to the ED(50) in the GFRalpha1 binding assay. The fusion protein was active in vivo in a rat middle cerebral artery occlusion model, where the stroke volume was reduced 77% (P < 0.001). In conclusion, these studies describe the re-engineering of GDNF, to make this neurotrophin transportable across the human BBB.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Delivery Systems/methods , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Animals , Glial Cell Line-Derived Neurotrophic Factor/genetics , Infarction, Middle Cerebral Artery/pathology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...