Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Hum Gene Ther ; 33(1-2): 37-60, 2022 01.
Article in English | MEDLINE | ID: mdl-34806402

ABSTRACT

Huntington's disease is a fatal neurodegenerative disorder caused by an inherited mutation in the huntingtin (HTT) gene comprising an expanded cytosine-adenine-guanine (CAG) trinucleotide repeat sequence that results in a pathogenic huntingtin protein. Adeno-associated viral (AAV) gene therapy containing a primary artificial microRNA (pri-amiRNA) specifically targeting HTT messenger RNA (mRNA) has the potential to provide long-lasting therapeutic benefit, through durable reduction of mutant HTT expression after a single administration. The efficiency and precision of processing of the pri-amiRNA precursor to the mature guide (G) strand by transduced cells are critical for specific and potent HTT mRNA lowering. The selection of the optimized pri-amiRNA comprised a series of in vitro studies followed by in vivo studies in small and then large mammals. Our studies demonstrate the predictivity of certain cell culture systems and rodent models for nonhuman primates with respect to some, but not all key features of pri-amiRNA processing. In addition, our results show that the processing of pri-amiRNAs to the mature guide strand can differ greatly across different scaffolds and sequences while providing the same levels of target lowering. Importantly, our data demonstrate that there is a combinatorial effect of guide and passenger (P) strand sequences, together with the scaffold, on pri-amiRNA processing, with different guide and passenger strand sequences within the same scaffold dramatically altering pri-amiRNA processing. Taken together, our results highlight the importance of optimizing not only target lowering but also the efficiency and precision of pri-amiRNA processing in vitro, in rodents and in large mammals to identify the most potent and selective AAV gene therapy that harnesses the endogenous microRNA (miRNA) biogenesis pathway for target lowering without perturbing the endogenous cellular miRNA profile. The optimized pri-amiRNA was selected with this focus on efficiency and precision of pri-amiRNA processing in addition to its pharmacological activity on HTT mRNA lowering and general tolerability in vivo.


Subject(s)
Huntington Disease , MicroRNAs , Animals , Genetic Therapy , Genetic Vectors/genetics , Huntingtin Protein/genetics , Huntington Disease/genetics , Huntington Disease/therapy , Mice , MicroRNAs/genetics , Primates/genetics
2.
Int J Toxicol ; 40(1): 4-14, 2021.
Article in English | MEDLINE | ID: mdl-33131343

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) is a potent neuroprotective biologic in Parkinson's disease models. Adeno-associated viral vector serotype 2 (AAV2)-human GDNF safety was assessed in rats treated with a single intracerebral dose of vehicle, 6.8 × 108, 6.8 × 109, or 5.2 × 1010 vector genomes (vg)/dose followed by interim sacrifices on day 7, 31, 90, and 376. There were no treatment-related effects observed on food consumption, body weight, hematology, clinical chemistry, coagulation parameters, neurobehavioral parameters, organ weights, or serum GDNF and anti-GDNF antibody levels. Increased serum anti-AAV2 neutralizing antibody titers were observed in the 5.2 × 1010 vg/dose group. Histopathological lesions were observed at the injection site in the 6.8 × 109 vg/dose (day 7) and 5.2 × 1010 vg/dose groups (days 7 and 31) and consisted of gliosis, mononuclear perivascular cuffing, intranuclear inclusion bodies, and/or apoptosis on day 7 and mononuclear perivascular cuffing on day 31. GDNF immunostaining was observed in the injection site in all dose groups through day 376 indicating no detectable impacts of anti-AAV2 neutralizing antibody. There was no evidence of increased expression of calcitonin gene-related peptide or Swann cell hyperplasia in the cervical and lumbar spinal cord or medulla oblongata at the 5.2 × 1010 vg/dose level indicating lack of hyperplastic effects. In conclusion, no systemic toxicity was observed, and the local toxicity observed at the injection site appeared to be reversible demonstrating a promising safety profile of intracerebral AAV2-GDNF delivery. Furthermore, an intracerebral dose of 6.8 × 108 AAV2-GDNF vg/dose was considered to be a no observed adverse effect level in rats.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/toxicity , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/toxicity , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Animals , Disease Models, Animal , Female , Humans , Male , Rats , Rats, Sprague-Dawley
3.
J Neurol Neurosurg Psychiatry ; 91(11): 1210-1218, 2020 11.
Article in English | MEDLINE | ID: mdl-32732384

ABSTRACT

Loss of nigrostriatal dopaminergic projection neurons is a key pathology in Parkinson's disease, leading to abnormal function of basal ganglia motor circuits and the accompanying characteristic motor features. A number of intraparenchymally delivered gene therapies designed to modify underlying disease and/or improve clinical symptoms have shown promise in preclinical studies and subsequently were evaluated in clinical trials. Here we review the challenges with surgical delivery of gene therapy vectors that limited therapeutic outcomes in these trials, particularly the lack of real-time monitoring of vector administration. These challenges have recently been addressed during the evolution of novel techniques for vector delivery that include the use of intraoperative MRI. The preclinical development of these techniques are described in relation to recent clinical translation in an adeno-associated virus serotype 2-mediated human aromatic L-amino acid decarboxylase gene therapy development programme. This new paradigm allows visualisation of the accuracy and adequacy of viral vector delivery within target structures, enabling intertrial modifications in surgical approaches, cannula design, vector volumes and dosing. The rapid, data-driven evolution of these procedures is unique and has led to improved vector delivery.


Subject(s)
Corpus Striatum , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Magnetic Resonance Imaging , Neurosurgical Procedures/methods , Parkinson Disease/therapy , Substantia Nigra , Animals , Aromatic-L-Amino-Acid Decarboxylases/genetics , Basal Ganglia , Dependovirus , Evidence-Based Medicine , GTP Cyclohydrolase/genetics , Glutamate Decarboxylase/genetics , Humans , Intraoperative Care/methods , Lentivirus , Neurturin/genetics , Parvovirinae , Primates , Surgery, Computer-Assisted , Tyrosine 3-Monooxygenase/genetics
4.
J Parkinsons Dis ; 10(3): 875-891, 2020.
Article in English | MEDLINE | ID: mdl-32508331

ABSTRACT

The concept of repairing the brain with growth factors has been pursued for many years in a variety of neurodegenerative diseases including primarily Parkinson's disease (PD) using glial cell line-derived neurotrophic factor (GDNF). This neurotrophic factor was discovered in 1993 and shown to have selective effects on promoting survival and regeneration of certain populations of neurons including the dopaminergic nigrostriatal pathway. These observations led to a series of clinical trials in PD patients including using infusions or gene delivery of GDNF or the related growth factor, neurturin (NRTN). Initial studies, some of which were open label, suggested that this approach could be of value in PD when the agent was injected into the putamen rather than the cerebral ventricles. In subsequent double-blind, placebo-controlled trials, the most recent reporting in 2019, treatment with GDNF did not achieve its primary end point. As a result, there has been uncertainty as to whether GDNF (and by extrapolation, related GDNF family neurotrophic factors) has merit in the future treatment of PD. To critically appraise the existing work and its future, a special workshop was held to discuss and debate this issue. This paper is a summary of that meeting with recommendations on whether there is a future for this therapeutic approach and also what any future PD trial involving GDNF and other GDNF family neurotrophic factors should consider in its design.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/metabolism , Neuroprotective Agents/therapeutic use , Parkinson Disease/therapy , Animals , Dopaminergic Neurons/metabolism , Genetic Therapy/methods , Glial Cell Line-Derived Neurotrophic Factor/genetics , Humans , Parkinson Disease/metabolism
5.
Ann Neurol ; 85(5): 704-714, 2019 05.
Article in English | MEDLINE | ID: mdl-30802998

ABSTRACT

OBJECTIVE: To understand the safety, putaminal coverage, and enzyme expression of adeno-associated viral vector serotype-2 encoding the complementary DNA for the enzyme, aromatic L-amino acid decarboxylase (VY-AADC01), delivered using novel intraoperative monitoring to optimize delivery. METHODS: Fifteen subjects (three cohorts of 5) with moderately advanced Parkinson's disease and medically refractory motor fluctuations received VY-AADC01 bilaterally coadministered with gadoteridol to the putamen using intraoperative magnetic resonance imaging (MRI) guidance to visualize the anatomic spread of the infusate and calculate coverage. Cohort 1 received 8.3 × 1011 vg/ml and ≤450 µl per putamen (total dose, ≤7.5 × 1011 vg); cohort 2 received the same concentration (8.3 × 1011 vg/ml) and ≤900 µl per putamen (total dose, ≤1.5 × 1012 vg); and cohort 3 received 2.6 × 1012 vg/ml and ≤900 µl per putamen (total dose, ≤4.7 × 1012 vg). (18)F-fluoro-L-dihydroxyphenylalanine positron emission tomography (PET) at baseline and 6 months postprocedure assessed enzyme activity; standard assessments measured clinical outcomes. RESULTS: MRI-guided administration of ascending VY-AADC01 doses resulted in putaminal coverage of 21% (cohort 1), 34% (cohort 2), and 42% (cohort 3). Cohorts 1, 2, and 3 showed corresponding increases in enzyme activity assessed by PET of 13%, 56%, and 79%, and reductions in antiparkinsonian medication of -15%, -33%, and -42%, respectively, at 6 months. At 12 months, there were dose-related improvements in clinical outcomes, including increases in patient-reported ON-time without troublesome dyskinesia (1.6, 3.3, and 1.5 hours, respectively) and quality of life. INTERPRETATION: Novel intraoperative monitoring of administration facilitated targeted delivery of VY-AADC01 in this phase 1 study, which was well tolerated. Increases in enzyme expression and clinical improvements were dose dependent. ClinicalTrials.gov Identifier: NCT01973543 Ann Neurol 2019;85:704-714.


Subject(s)
Aromatic-L-Amino-Acid Decarboxylases/genetics , Genetic Therapy/methods , Magnetic Resonance Imaging/methods , Parkinson Disease/diagnostic imaging , Parkinson Disease/genetics , Putamen/diagnostic imaging , Adult , Aged , Aromatic-L-Amino-Acid Decarboxylases/administration & dosage , Female , Gene Transfer Techniques , Humans , Male , Middle Aged , Parkinson Disease/therapy
6.
J Neurosurg ; 132(2): 595-604, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30771782

ABSTRACT

OBJECTIVE: Successful convection-enhanced delivery of therapeutic agents to subcortical brain structures requires accurate cannula placement. Stereotactic guiding devices have been developed to accurately target brain nuclei. However, technologies remain limited by a lack of MRI compatibility, or by devices' size, making them suboptimal for direct gene delivery to brain parenchyma. The goal of this study was to validate the accuracy of a novel frameless skull-mounted ball-joint guide array (BJGA) in targeting the nonhuman primate (NHP) brain. METHODS: Fifteen MRI-guided cannula insertions were performed on 9 NHPs, each targeting the putamen. Optimal trajectories were planned on a standard MRI console using 3D multiplanar baseline images. After cannula insertion, the intended trajectory was compared to the final trajectory to assess deviation (euclidean error) of the cannula tip. RESULTS: The average cannula tip deviation was 1.18 ± 0.60 mm (mean ± SD) as measured by 2 independent reviewers. Topological analysis showed a superior, posterior, and rightward directional bias, and the intra- and interclass correlation coefficients were > 0.85, indicating valid and reliable intra- and interobserver evaluation. CONCLUSIONS: The data demonstrate that the BJGA can be used to reliably target subcortical brain structures by using MRI guidance, with accuracy comparable to current frameless stereotactic systems. The size and versatility of the BJGA, combined with a streamlined workflow, allows for its potential applicability to a variety of intracranial neurosurgical procedures, and for greater flexibility in executing MRI-guided experiments within the NHP brain.


Subject(s)
Magnetic Resonance Imaging/methods , Neuronavigation/methods , Neurosurgical Procedures/methods , Skull/diagnostic imaging , Skull/surgery , Animals , Brain/diagnostic imaging , Brain/surgery , Macaca mulatta , Magnetic Resonance Imaging/instrumentation , Male , Neuronavigation/instrumentation , Neurosurgical Procedures/instrumentation , Stereotaxic Techniques/instrumentation
7.
J Neurosci Methods ; 293: 347-358, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29042259

ABSTRACT

BACKGROUND: In non-human primate (NHP) optogenetics, infecting large cortical areas with viral vectors is often a difficult and time-consuming task. Previous work has shown that parenchymal delivery of adeno-associated virus (AAV) in the thalamus by convection-enhanced delivery (CED) can lead to large-scale transduction via axonal transport in distal areas including cortex. We used this approach to obtain widespread cortical expression of light-sensitive ion channels. NEW METHOD: AAV vectors co-expressing channelrhodopsin-2 (ChR2) and yellow fluorescent protein (YFP) genes were infused into thalamus of three rhesus macaques under MR-guided CED. After six to twelve weeks recovery, in vivo optical stimulation and single cell recording in the cortex was carried out using an optrode in anesthetized animals. Post-mortem immunostaining against YFP was used to estimate the distribution and level of expression of ChR2 in thalamus and cortex. RESULTS: Histological analysis revealed high levels of transduction in cortical layers. The patterns of expression were consistent with known thalamo-cortico-thalamic circuits. Dense expression was seen in thalamocortiocal axonal fibers in layers III, IV and VI and in pyramidal neurons in layers V and VI, presumably corticothalamic neurons. In addition we obtained reliable in vivo light-evoked responses in cortical areas with high levels of expression. COMPARISON WITH EXISTING METHODS: Thalamic CED is very efficient in achieving large expressing areas in comparison to convectional techniques both in minimizing infusion time and in minimizing damage to the brain. CONCLUSION: MR-guided CED infusion into thalamus provides a simplified approach to transduce large cortical areas by thalamo-cortico-thalamic projections in primate brain.


Subject(s)
Dependovirus/genetics , Genetic Vectors/administration & dosage , Macaca mulatta , Optogenetics/methods , Thalamus , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/physiology , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Convection , Dermoscopy , Female , Imaging, Three-Dimensional , Immunohistochemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Magnetic Resonance Imaging , Male , Models, Animal , Neural Pathways/cytology , Neural Pathways/physiology , Photic Stimulation , Thalamus/cytology , Thalamus/diagnostic imaging , Thalamus/physiology
8.
Addict Biol ; 20(4): 629-42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-24801661

ABSTRACT

Moderate social consumption of alcohol is common; however, only a small percentage of individuals transit from social to excessive, uncontrolled alcohol drinking. This suggests the existence of protective mechanisms that prevent the development of alcohol addiction. Here, we tested the hypothesis that the glial cell line-derived neurotrophic factor (GDNF) in the mesolimbic system [e.g. the nucleus accumbens (Acb) and ventral tegmental area (VTA)] is part of such a mechanism. We found that GDNF knockdown, by infecting rat Acb neurons with a small hairpin RNA (shRNA) targeting the GDNF gene, produced a rapid escalation to excessive alcohol consumption and enhanced relapse to alcohol drinking. Conversely, viral-mediated overexpression of the growth factor in the mesolimbic system blocked the escalation from moderate to excessive alcohol drinking. To access the mechanism underlying GDNF's actions, we measured the firing rate of dopaminergic (DAergic) neurons in the VTA after a history of excessive alcohol intake with or without elevating GDNF levels. We found that the spontaneous firing rate of DAergic neurons in the VTA was reduced during alcohol withdrawal and that GDNF reversed this alcohol-induced DA deficiency. Together, our results suggest that endogenous GDNF in the mesolimbic system controls the transition from moderate to excessive alcohol drinking and relapse via reversal of alcohol-dependent neuro-adaptations in DAergic VTA neurons.


Subject(s)
Alcohol Drinking/physiopathology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Limbic System/physiology , Nucleus Accumbens/physiology , Ventral Tegmental Area/physiology , Adaptation, Physiological/physiology , Animals , Conditioning, Operant , Dopaminergic Neurons/physiology , Down-Regulation/physiology , Gene Knockdown Techniques , Glial Cell Line-Derived Neurotrophic Factor/deficiency , Male , Rats, Long-Evans , Recurrence , Self Administration , Up-Regulation/physiology
9.
Article in English | MEDLINE | ID: mdl-25541617

ABSTRACT

Aromatic L-amino acid decarboxylase (AADC) deficiency is a rare, autosomal-recessive neurological disorder caused by mutations in the DDC gene that leads to an inability to synthesize catecholamines and serotonin. As a result, patients suffer compromised development, particularly in motor function. A recent gene replacement clinical trial explored putaminal delivery of recombinant adeno-associated virus serotype 2 vector encoding human AADC (AAV2-hAADC) in AADC-deficient children. Unfortunately, patients presented only modest amelioration of motor symptoms, which authors acknowledged could be due to insufficient transduction of putamen. We hypothesize that, with the development of a highly accurate MRI-guided cannula placement technology, a more effective approach might be to target the affected mid-brain neurons directly. Transduction of AADC-deficient dopaminergic neurons in the substantia nigra and ventral tegmental area with locally infused AAV2-hAADC would be expected to lead to restoration of normal dopamine levels in affected children. The objective of this study was to assess the long-term safety and tolerability of bilateral AAV2-hAADC MRI-guided pressurized infusion into the mid-brain of non-human primates. Animals received either vehicle, low or high AAV2-hAADC vector dose and were euthanized 1, 3 or 9 months after surgery. Our data indicate that effective mid-brain transduction was achieved without untoward effects.

10.
Hum Gene Ther ; 25(7): 619-30, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24617515

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disease caused by mutations in survival motor neuron 1 (SMN1). Previously, we showed that central nervous system (CNS) delivery of an adeno-associated viral (AAV) vector encoding SMN1 produced significant improvements in survival in a mouse model of SMA. Here, we performed a dose-response study in SMA mice to determine the levels of SMN in the spinal cord necessary for efficacy, and measured the efficiency of motor neuron transduction in the spinal cord after intrathecal delivery in pigs and nonhuman primates (NHPs). CNS injections of 5e10, 1e10, and 1e9 genome copies (gc) of self-complementary AAV9 (scAAV9)-hSMN1 into SMA mice extended their survival from 17 to 153, 70, and 18 days, respectively. Spinal cords treated with 5e10, 1e10, and 1e9 gc showed that 70-170%, 30-100%, and 10-20% of wild-type levels of SMN were attained, respectively. Furthermore, detectable SMN expression in a minimum of 30% motor neurons correlated with efficacy. A comprehensive analysis showed that intrathecal delivery of 2.5e13 gc of scAAV9-GFP transduced 25-75% of the spinal cord motor neurons in NHPs. Thus, the extent of gene expression in motor neurons necessary to confer efficacy in SMA mice could be obtained in large-animal models, justifying the continual development of gene therapy for SMA.


Subject(s)
Dependovirus , Genetic Vectors/pharmacology , Injections, Spinal , Muscular Atrophy, Spinal/therapy , Protein Biosynthesis , Survival of Motor Neuron 1 Protein , Animals , Genetic Vectors/genetics , Genetic Vectors/metabolism , Mice , Mice, Knockout , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Survival of Motor Neuron 1 Protein/biosynthesis , Survival of Motor Neuron 1 Protein/genetics , Swine
11.
Front Neuroanat ; 8: 9, 2014.
Article in English | MEDLINE | ID: mdl-24672434

ABSTRACT

When nanoparticles/proteins are infused into the brain, they are often transported to distal sites in a manner that is dependent both on the characteristics of the infusate and the region targeted. We have previously shown that adeno-associated virus (AAV) is disseminated within the brain by perivascular flow and also by axonal transport. Perivascular distribution usually does not depend strongly on the nature of the infusate. Many proteins, neutral liposomes and AAV particles distribute equally well by this route when infused under pressure into various parenchymal locations. In contrast, axonal transport requires receptor-mediated uptake of AAV by neurons and engagement with specific transport mechanisms previously demonstrated for other neurotropic viruses. Cerebrospinal fluid (CSF) represents yet another way in which brain anatomy may be exploited to distribute nanoparticles broadly in the central nervous system. In this study, we assessed the distribution and perivascular transport of nanoparticles of different sizes delivered into the parenchyma of rodents and CSF in non-human primates.

12.
Mol Ther ; 22(2): 329-337, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24419081

ABSTRACT

Many studies have demonstrated that adeno-associated virus serotype 9 (AAV9) transduces astrocytes and neurons when infused into rat or nonhuman primate (NHP) brain. We previously showed in rats that transduction of antigen-presenting cells (APC) by AAV9 encoding a foreign protein triggered a full neurotoxic immune response. Accordingly, we asked whether this phenomenon occurred in NHP. We performed parenchymal or intrathecal infusion of AAV9 encoding green fluorescent protein (GFP), a non-self protein derived from jellyfish, or human aromatic L-amino acid decarboxylase (hAADC), a self-protein, in separate NHP. Animals receiving AAV9-GFP into cisterna magna (CM) became ataxic, indicating cerebellar pathology, whereas AAV9-hAADC animals remained healthy. In transduced regions, AAV9-GFP elicited inflammation associated with early activation of astrocytic and microglial cells, along with upregulation of major histocompatibility complex class II (MHC-II) in glia. In addition, we found Purkinje neurons lacking calbindin after AAV9-GFP but not after AAV9-hAADC delivery. Our results demonstrate that AAV9-mediated expression of a foreign-protein, but not self-recognized protein, triggers complete immune responses in NHP regardless of the route of administration. Our results warrant caution when contemplating use of serotypes that can transduce APC if the transgene is not syngeneic with the host. This finding has the potential to complicate preclinical toxicology studies in which such vectors encoding human cDNA's are tested in animals.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Central Nervous System/immunology , Central Nervous System/metabolism , Dependovirus , Genetic Vectors , Inflammation/genetics , Inflammation/immunology , Animals , Central Nervous System/pathology , Corpus Striatum/immunology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dependovirus/genetics , Dependovirus/immunology , Gene Expression , Genes, Reporter , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/immunology , Green Fluorescent Proteins/genetics , Humans , Inflammation/pathology , Neurons/metabolism , Neurons/pathology , Rats , Transduction, Genetic , Transgenes
13.
Magn Reson Med ; 71(1): 19-25, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24346964

ABSTRACT

PURPOSE: To investigate hyperpolarized (13) C metabolic imaging methods in the primate brain that can be translated into future clinical trials for patients with brain cancer. METHODS: (13) C coils and pulse sequences designed for use in humans were tested in phantoms. Dynamic (13) C data were obtained from a healthy cynomolgus monkey brain using the optimized (13) C coils and pulse sequences. The metabolite kinetics were estimated from two-dimensional localized (13) C dynamic imaging data from the nonhuman primate brain. RESULTS: Pyruvate and lactate signal were observed in both the brain and the surrounding tissues with the maximum signal-to-noise ratio of 218 and 29 for pyruvate and lactate, respectively. Apparent rate constants for the conversion of pyruvate to lactate and the ratio of lactate to pyruvate showed a difference between brain and surrounding tissues. CONCLUSION: The feasibility of using hyperpolarized [1-(13) C]-pyruvate for assessing in vivo metabolism in a healthy nonhuman primate brain was demonstrated using a hyperpolarized (13) C imaging experimental setup designed for studying patients with brain tumors. The kinetics of the metabolite conversion suggests that this approach may be useful in future studies of human neuropathology.


Subject(s)
Brain/metabolism , Lactic Acid/metabolism , Magnetic Resonance Imaging/instrumentation , Magnetic Resonance Spectroscopy/instrumentation , Pyruvic Acid/metabolism , Animals , Brain/anatomy & histology , Carbon Isotopes/pharmacokinetics , Equipment Design , Equipment Failure Analysis , Feasibility Studies , Female , Humans , Macaca fascicularis , Magnetic Resonance Imaging/methods , Magnetic Resonance Spectroscopy/methods , Reproducibility of Results , Sensitivity and Specificity
14.
PLoS One ; 8(6): e64452, 2013.
Article in English | MEDLINE | ID: mdl-23755125

ABSTRACT

This study presents a computational tool for auto-segmenting the distribution of brain infusions observed by magnetic resonance imaging. Clinical usage of direct infusion is increasing as physicians recognize the need to attain high drug concentrations in the target structure with minimal off-target exposure. By co-infusing a Gadolinium-based contrast agent and visualizing the distribution using real-time using magnetic resonance imaging, physicians can make informed decisions about when to stop or adjust the infusion. However, manual segmentation of the images is tedious and affected by subjective preferences for window levels, image interpolation and personal biases about where to delineate the edge of the sloped shoulder of the infusion. This study presents a computational technique that uses a Gaussian Mixture Model to efficiently classify pixels as belonging to either the high-intensity infusate or low-intensity background. The algorithm was implemented as a distributable plug-in for the widely used imaging platform OsiriX®. Four independent operators segmented fourteen anonymized datasets to validate the tool's performance. The datasets were intra-operative magnetic resonance images of infusions into the thalamus or putamen of non-human primates. The tool effectively reproduced the manual segmentation volumes, while significantly reducing intra-operator variability by 67±18%. The tool will be used to increase efficiency and reduce variability in upcoming clinical trials in neuro-oncology and gene therapy.


Subject(s)
Contrast Media/pharmacokinetics , Heterocyclic Compounds/pharmacokinetics , Image Interpretation, Computer-Assisted , Neuroimaging , Organometallic Compounds/pharmacokinetics , Software , Algorithms , Animals , Contrast Media/administration & dosage , Dependovirus/genetics , Gadolinium/administration & dosage , Gadolinium/pharmacokinetics , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/pharmacokinetics , Heterocyclic Compounds/administration & dosage , Infusions, Intraventricular , Magnetic Resonance Imaging , Observer Variation , Organometallic Compounds/administration & dosage , Primates , Putamen/metabolism , Thalamus/metabolism
15.
Neurotherapeutics ; 10(3): 498-510, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23700209

ABSTRACT

Protein aggregation as a result of misfolding is a common theme underlying neurodegenerative diseases. Accordingly, most recent studies aim to prevent protein misfolding and/or aggregation as a strategy to treat these pathologies. For instance, state-of-the-art approaches, such as silencing protein overexpression by means of RNA interference, are being tested with positive outcomes in preclinical models of animals overexpressing the corresponding protein. Therapies designed to treat central nervous system diseases should provide accurate delivery of the therapeutic agent and long-term or chronic expression by means of a nontoxic delivery vehicle. After several years of technical advances and optimization, gene therapy emerges as a promising approach able to fulfill those requirements. In this review we will summarize the latest improvements achieved in gene therapy for central nervous system diseases associated with protein misfolding (e.g., amyotrophic lateral sclerosis, Alzheimer's, Parkinson's, Huntington's, and prion diseases), as well as the most recent approaches in this field to treat these pathologies.


Subject(s)
Central Nervous System Diseases , Genetic Therapy/methods , Proteostasis Deficiencies , Animals , Central Nervous System Diseases/complications , Central Nervous System Diseases/genetics , Central Nervous System Diseases/therapy , Humans , Proteostasis Deficiencies/complications , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/therapy
16.
Hum Gene Ther ; 24(5): 526-32, 2013 May.
Article in English | MEDLINE | ID: mdl-23517473

ABSTRACT

The present study builds on previous work showing that infusion of adeno-associated virus type 9 (AAV9) into the cisterna magna (CM) of nonhuman primates resulted in widespread transduction throughout cortex and spinal cord. Transduction efficiency was severely limited, however, by the presence of circulating anti-AAV antibodies. Accordingly, we compared AAV9 to a related serotype, AAV7, which has a high capsid homology. CM infusion of either AAV7 or AAV9 directed high level of cell transduction with similar patterns of distribution throughout brain cortex and along the spinal cord. Dorsal root ganglia and corticospinal tracts were also transduced. Both astrocytes and neurons were transduced. Interestingly, little transduction was observed in peripheral organs. Our results indicate that intrathecal delivery of either AAV7 or AAV9 directs a robust and widespread cellular transduction in the central nervous system and other peripheral neural structures.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques/adverse effects , Genetic Vectors/adverse effects , Macaca/genetics , Transduction, Genetic , Animals , Astrocytes/pathology , Astrocytes/virology , Cerebral Cortex/pathology , Cerebral Cortex/virology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Ganglia, Spinal/virology , Genetic Vectors/cerebrospinal fluid , Green Fluorescent Proteins/genetics , Macaca/virology , Motor Neurons/metabolism , Motor Neurons/pathology , Motor Neurons/virology , Spinal Cord/pathology , Spinal Cord/virology
17.
J Neurosci Methods ; 213(2): 214-27, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23313849

ABSTRACT

In 1873 Camillo Golgi discovered a staining technique that allowed for the visualization of whole neurons within the brain, initially termed 'the black reaction' and is now known as Golgi impregnation. Despite the capricious nature of this method, Golgi impregnation remains a widely used method for whole neuron visualization and analysis of dendritic arborization and spine quantification. We describe a series of reliable, modified 'Golgi-Cox' impregnation methods that complement some existing methods and have several advantages over traditional whole brain 'Golgi' impregnation. First, these methods utilize 60-100µm thick brain sections, which allows for fast, reliable impregnation of neurons in rats (7-14 days) and non-human primates (NHP) (30 days) while avoiding the pitfalls of other 'rapid Golgi' techniques traditionally employed with thin sections. Second, these methods employ several common tissue fixatives, resulting in high quality neuron impregnation in brain sections from acrolein, glutaraldehyde, and paraformaldehyde perfused rats, and in glutaraldehyde perfused NHP brain tissue. Third, because thin sections are obtained on a vibratome prior to processing, alternate sections of brain tissue can be used for additional analyses such as immunohistochemistry or electron microscopy. This later advantage allows for comparison of, for example, dendrite morphology in sections adjacent to pertinent histochemical markers or ultrastructural components. Finally, we describe a method for simultaneous light microscopic visualization of both tyrosine hydroxylase immunohistochemistry and Golgi impregnation in the same tissue section. Thus, the methods described here allow for fast, high quality Golgi impregnation and conserve experimental subjects by allowing multiple analyses within an individual animal.


Subject(s)
Brain/ultrastructure , Neurons/ultrastructure , Staining and Labeling/methods , Tissue Fixation/methods , Animals , Macaca mulatta , Male , Microscopy, Immunoelectron , Rats , Rats, Sprague-Dawley
18.
Hum Gene Ther ; 23(8): 891-902, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22574943

ABSTRACT

Niemann-Pick disease is a lysosomal storage disorder resulting from inherited deficiency in acid sphingomyelinase (ASM). Use of adeno-associated virus serotype 2 (AAV2) to deliver human acid sphingomyelinase (hASM) is currently being explored as a means to treat the devastating neurological features of NPD, which are refractory to traditional enzyme replacement therapy. In this study, we evaluated the long-term efficacy and safety of AAV2-hASM after direct infusion into the CNS of nonhuman primates. First, we confirmed the efficacy of AAV2-hASM in naive rats, which exhibited increased ASM expression and enzyme activity after infusion, without evidence of local or systemic toxicity. Next, the model was adapted to naive nonhuman primates (NHPs) with various doses of AAV2-hASM or saline delivered into the brainstem and both thalami. Strikingly, NHPs that received a high dose of AAV2-hASM displayed significant motor deficits that were not seen in low-dose animals in both the short-term (3-month) and long-term (9-month) treatment groups. In treated NHPs, ASM expression and activity were elevated with associated alterations in the sphingolipidomic profile in brain regions transduced with AAV2-hASM. Initial histological analysis indicated marked inflammatory reactions, and immunohistochemical analysis confirmed a robust inflammatory response. Importantly, pronounced upregulation of the chemokine CCL5, a target of ASM-mediated inflammatory signaling, was detected that correlated with the inflammatory response, providing a possible mechanism for hASM-associated toxicity. This study defines dose-dependent and dose-independent toxicities of AAV2-hASM in the naive primate brain, and reveals potential challenges in the design of a clinical trial.


Subject(s)
Brain/metabolism , Brain/pathology , Dependovirus/genetics , Sphingomyelin Phosphodiesterase/genetics , Animals , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Genetic Therapy , Genetic Vectors , Humans , Niemann-Pick Diseases/therapy , Rats , Rats, Sprague-Dawley , Sphingomyelin Phosphodiesterase/metabolism , Up-Regulation
19.
Hum Gene Ther ; 23(2): 210-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22017504

ABSTRACT

Degeneration of nigrostriatal neurons in Parkinson's disease (PD) causes progressive loss of aromatic l-amino acid decarboxylase (AADC), the enzyme that converts levodopa (l-DOPA) into dopamine in the striatum. Because loss of this enzyme appears to be a major driver of progressive impairment of response to the mainstay drug, l-DOPA, one promising approach has been to use gene therapy to restore AADC activity in the human putamen and thereby restore normal l-DOPA response in patients with PD. An open-label phase I clinical trial of this approach in patients with PD provided encouraging signs of improvement in Unified Parkinson's Disease Rating Scale scores and reductions in antiparkinsonian medications. However, such improvement was modest compared with the results previously reported in parkinsonian rhesus macaques. The reason for this discrepancy may have been that the relatively small volume of vector infused in the clinical study restricted the distribution of AADC expression, such that only about 20% of the postcommissural putamen was covered, as revealed by l-[3-(18)F]-α-methyltyrosine-positron emission tomography. To achieve more quantitative distribution of vector, we have developed a visual guidance system for parenchymal infusion of AAV2. The purpose of the present study was to evaluate the combined magnetic resonance imaging-guided delivery system with AAV2-hAADC under conditions that approximate the intended clinical protocol. Our data indicate that this approach directed accurate cannula placement and effective vector distribution without inducing any untoward effects in nonhuman primates infused with a high dose of AAV2-hAADC.


Subject(s)
Corpus Striatum/enzymology , Dependovirus/genetics , Dopa Decarboxylase/genetics , Gene Transfer Techniques , Animals , Catheterization , Caudate Nucleus/enzymology , Dopa Decarboxylase/metabolism , Female , Humans , Macaca mulatta , Magnetic Resonance Imaging , Neurons/enzymology , Neurons/pathology , Putamen/enzymology , Putamen/pathology , Stereotaxic Techniques , Transgenes
20.
Neurobiol Dis ; 48(2): 228-35, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22019719

ABSTRACT

Delivery of neurotrophic factors to treat neurodegenerative diseases has not been efficacious in clinical trials despite their known potency for promoting neuronal growth and survival. Direct gene delivery to the brain offers an approach for establishing sustained expression of neurotrophic factors but is dependent on accurate surgical procedures to target specific anatomical regions of the brain. Serotype-2 adeno-associated viral (AAV2) vectors have been investigated in multiple clinical studies for neurological diseases without adverse effects; however the absence of significant clinical efficacy after neurotrophic factor gene transfer has been largely attributed to insufficient coverage of the target region. Our pre-clinical development of AAV2-glial-derived neurotrophic factor (GDNF) for Parkinson's disease involved real-time image guided delivery and optimization of delivery techniques to maximize gene transfer in the putamen. We have demonstrated that AAV2 vectors are anterogradely transported in the primate brain with GDNF expression observed in the substantia nigra after putaminal delivery in both intact and nigrostriatal lesioned primates. Direct midbrain delivery of AAV2-GDNF resulted in extensive anterograde transport to multiple brain regions and significant weight loss.


Subject(s)
Brain/virology , Dependovirus/genetics , Genetic Therapy/methods , Glial Cell Line-Derived Neurotrophic Factor/genetics , Parkinson Disease/therapy , Animals , Brain Chemistry , Drug Delivery Systems , Gene Transfer Techniques , Genetic Vectors , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Primates
SELECTION OF CITATIONS
SEARCH DETAIL
...