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










Publication year range
1.
Mol Ther Methods Clin Dev ; 20: 389-397, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33575431

ABSTRACT

Infantile malignant osteopetrosis is a devastating disorder of early childhood that is frequently fatal and for which there are only limited therapeutic options. Gene therapy utilizing autologous hematopoietic stem and progenitor cells represents a potentially advantageous therapeutic alternative for this multisystemic disease. Gene therapy can be performed relatively rapidly following diagnosis, will not result in graft versus host disease, and may also have potential for reduced incidences of other transplant-related complications. In this review, we have summarized the past sixteen years of research aimed at developing a gene therapy for infantile malignant osteopetrosis; these efforts have culminated in the first clinical trial employing lentiviral-mediated delivery of TCIRG1 in autologous hematopoietic stem and progenitor cells.

2.
Alzheimers Dement ; 10(5): 571-81, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24411134

ABSTRACT

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


Subject(s)
Alzheimer Disease/therapy , Dependovirus/genetics , Genetic Therapy/methods , Nerve Growth Factor/genetics , Aged , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Basal Nucleus of Meynert , Feasibility Studies , Female , Genetic Vectors , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Male , Middle Aged , Neuropsychological Tests , Neurosurgical Procedures , Positron-Emission Tomography , Stereotaxic Techniques , Treatment Outcome
3.
Neurobiol Dis ; 58: 38-48, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23631873

ABSTRACT

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


Subject(s)
Adenoviridae/genetics , Corpus Striatum/metabolism , Nerve Growth Factors/administration & dosage , Neurodegenerative Diseases/prevention & control , Neuroprotective Agents/administration & dosage , Substantia Nigra/metabolism , Adrenergic Agents/toxicity , Animals , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Genetic Vectors/physiology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Male , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , Neuroprotective Agents/metabolism , Oxidopamine/toxicity , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase
4.
Neurology ; 80(18): 1698-701, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23576625

ABSTRACT

OBJECTIVE: In an effort to account for deficiencies in axonal transport that limit the effectiveness of neurotrophic factors, this study tested the safety and feasibility, in moderately advanced Parkinson disease (PD), of bilaterally administering the gene therapy vector AAV2-neurturin (CERE-120) to the putamen plus substantia nigra (SN, a relatively small structure deep within the midbrain, in proximity to critical neuronal and vascular structures). METHODS: After planning and minimizing risks of stereotactically targeting the SN, an open-label, dose-escalation safety trial was initiated in 6 subjects with PD who received bilateral stereotactic injections of CERE-120 into the SN and putamen. RESULTS: Two-year safety data for all subjects suggest the procedures were well-tolerated, with no serious adverse events. All adverse events and complications were expected for patients with PD undergoing stereotactic brain surgery. CONCLUSIONS: Bilateral stereotactic administration of CERE-120 to the SN plus putamen in PD is feasible and this evaluation provides initial empirical support that it is safe and well-tolerated. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence that bilateral neurturin gene delivery (CERE-120) to the SN plus putamen in patients with moderately advanced PD is feasible and safe.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Neurturin/genetics , Parkinson Disease/therapy , Stereotaxic Techniques , Substantia Nigra/physiology , Feasibility Studies , Female , Follow-Up Studies , Gene Transfer Techniques/adverse effects , Genetic Therapy/adverse effects , Humans , Male , Microinjections/methods , Middle Aged , Treatment Outcome
5.
Neurobiol Aging ; 34(1): 35-61, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22926166

ABSTRACT

Neurotrophic factors have long shown promise as potential therapies for age-related neurodegenerative diseases. However, 20 years of largely disappointing clinical results have underscored the difficulties involved with safely and effectively delivering these proteins to targeted sites within the central nervous system. Recent progress establishes that gene transfer can now likely overcome the delivery issues plaguing the translation of neurotrophic factors. This may be best exemplified by adeno-associated virus serotype-2-neurturin (CERE-120), a viral-vector construct designed to deliver the neurotrophic factor, neurturin to degenerating nigrostriatal neurons in Parkinson's disease. Eighty Parkinson's subjects have been dosed with CERE-120 (some 7+ years ago), with long-term, targeted neurturin expression confirmed and no serious safety issues identified. A double-blind, controlled Phase 2a trial established clinical "proof-of-concept" via 19 of the 24 prescribed efficacy end points favoring CERE-120 at the 12-month protocol-prescribed time point and all but one favoring CERE-120 at the 18-month secondary time point (p = 0.007 and 0.001, respectively). Moreover, clinically meaningful benefit was seen with CERE-120 on several specific protocol-prescribed, pairwise, blinded, motor, and quality-of-life end points at 12 months, and an even greater number of end points at 18 months. Because the trial failed to meet the primary end point (Unified Parkinson's Disease Rating Scale motor-off, measured at 12 months), a revised multicenter Phase 1/2b protocol was designed to enhance the neurotrophic effects of CERE-120, using insight gained from the Phase 2a trial. This review summarizes the development of CERE-120 from its inception through establishing "clinical proof-of-concept" and beyond. The translational obstacles and issues confronted, and the strategies applied, are reviewed. This information should be informative to investigators interested in translational research and development for age-related and other neurodegenerative diseases.


Subject(s)
Aging , Genetic Therapy , Neurodegenerative Diseases/therapy , Neurturin/therapeutic use , Parkinson Disease/therapy , Animals , Genetic Vectors/physiology , Humans , Neurturin/biosynthesis , Neurturin/genetics
6.
Neurobiol Dis ; 44(1): 38-52, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21704161

ABSTRACT

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


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Neurturin/metabolism , Parkinson Disease/therapy , Substantia Nigra/metabolism , Animals , Behavior, Animal/physiology , Diet , Gene Dosage , Gene Targeting , Genetic Vectors , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor/genetics , Green Fluorescent Proteins , Immunohistochemistry , Male , Neurons/metabolism , Neurturin/adverse effects , Rats , Rats, Sprague-Dawley , Risk Assessment , Tyrosine 3-Monooxygenase/metabolism , Weight Gain/genetics , Weight Gain/physiology , Weight Loss/physiology
7.
Mov Disord ; 26(1): 27-36, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21322017

ABSTRACT

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


Subject(s)
Corpus Striatum/metabolism , Genetic Therapy/methods , MPTP Poisoning/therapy , Neurturin/therapeutic use , Parkinson Disease/therapy , Aged , Animals , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Functional Laterality , Gene Expression Regulation/drug effects , Humans , MPTP Poisoning/chemically induced , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Macaca mulatta , Male , Middle Aged , Neurturin/genetics , Neurturin/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Tyrosine 3-Monooxygenase/metabolism
8.
Drug Deliv Transl Res ; 1(5): 361-82, 2011 Oct.
Article in English | MEDLINE | ID: mdl-25788422

ABSTRACT

Efforts to develop neurotrophic factors to restore function and protect dying neurons in chronic neurodegenerative diseases like Alzheimer's (AD) and Parkinson's (PD) have been attempted for decades. Despite abundant data establishing nonclinical proof-of-concept, significant delivery issues have precluded the successful translation of this concept to the clinic. The development of AAV2 viral vectors to deliver therapeutic genes has emerged as a safe and effective means to achieve sustained, long-term, targeted, bioactive protein expression. Thus, it potentially offers a practical means to solve those long-standing delivery/translational issues associated with neurotrophic factors. Data are presented for two AAV2 viral vector constructs expressing one of two different neurotrophic factors: nerve growth factor (NGF) and neurturin (NRTN). One (AAV2-NGF; aka CERE-110) is being developed as a treatment to improve the function and delay further degeneration of cholinergic neurons in the nucleus basalis of Meynert, the degeneration of which has been linked to cognitive deficits in AD. The other (AAV2-NRTN; aka CERE-120) is similarly being developed to treat the degenerating nigrostriatal dopamine neurons and major motor deficits in PD. The data presented here demonstrate: (1) 2-year, targeted, bioactive-protein in monkeys, (2) persistent, bioactive-protein throughout the life-span of the rat, and (3) accurately targeted bioactive-protein in aged rats, with (4) no safety issues or antibodies to the protein detected. They also provide empirical guidance to establish parameters for human dosing and collectively support the idea that gene transfer may overcome key delivery obstacles that have precluded successful translation of neurotrophic factors to the clinic. More specifically, they also enabled the AAV-NGF and AAV-NRTN programs to advance into ongoing multi-center, double-blind clinical trials in AD and PD patients.

9.
Neurosurgery ; 64(4): 602-12; discussion 612-3, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19349823

ABSTRACT

OBJECTIVE: Parkinson's disease is characterized by profound motor deficits that result mainly as a consequence of degeneration of midbrain dopaminergic neurons. No current therapy slows or halts disease progression. Neurturin (NTN) and glial cell line-derived neurotrophic factor have potent neuroprotective and neurorestorative effects on dopaminergic neurons, but their use in treating Parkinson's disease has been limited by significant delivery obstacles. In this study, we examined the long-term expression, bioactivity, and safety/tolerability of CERE-120, an adeno-associated virus type 2 vector encoding human NTN, after bilateral stereotactic delivery to the striatum of nonhuman primates. METHODS: Twelve naïve rhesus macaques received bilateral stereotactic injections of 1 of 2 CERE-120 doses or vehicle to the caudate and putamen. Neurological and clinical parameters were monitored for up to 1 year postadministration, after which animals were sacrificed for histological analyses. RESULTS: Dose-related NTN expression was observed at 1 year and was associated with enhanced tyrosine hydroxylase immunolabeling in the striatum, hypertrophy of tyrosine hydroxylase-positive cells in the substantia nigra, and induction of extracellular signal-regulated kinase signaling in the substantia nigra. Extensive, formal analyses, conducted in accordance with Good Laboratory Practice Regulations, across multiple time points revealed no evidence of clinical, neurological, or systemic toxicity. CONCLUSION: The present study provides evidence of long-term expression and bioactivity of NTN on the dopaminergic nigrostriatal system after bilateral stereotactic delivery of CERE-120 to the striatum. Furthermore, no evidence of any adverse effects for up to 1 year postadministration was observed. These findings reveal a wide safety margin for CERE-120 and collectively support the ongoing clinical testing of the efficacy and safety of CERE-120 in patients with Parkinson's disease.


Subject(s)
Corpus Striatum/drug effects , Dependovirus/genetics , Gene Expression/physiology , Neurturin/administration & dosage , Neurturin/pharmacology , Parkinson Disease/drug therapy , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Behavior, Animal/drug effects , Behavior, Animal/physiology , Body Weight/drug effects , Brain/metabolism , Calcitonin Gene-Related Peptide/metabolism , Cell Count , Corpus Striatum/metabolism , Corpus Striatum/virology , Dependovirus/metabolism , Dose-Response Relationship, Drug , Eating/drug effects , Enzyme-Linked Immunosorbent Assay/methods , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Transfer Techniques , Genetic Vectors , Humans , Longitudinal Studies , Macaca mulatta , Neurturin/genetics , Neurturin/metabolism , Spinal Cord/metabolism , Time Factors , Tissue Distribution , Tyrosine 3-Monooxygenase/metabolism , Viral Matrix Proteins/metabolism
10.
Neurobiol Dis ; 34(1): 40-50, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19150499

ABSTRACT

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


Subject(s)
Cerebral Cortex/physiopathology , Corpus Striatum/physiopathology , Genetic Therapy , Huntington Disease/therapy , Motor Activity , Neurons/physiology , Neurturin/genetics , Animals , DNA-Binding Proteins , Dependovirus/genetics , Disease Models, Animal , Gene Expression , Gene Transfer Techniques , Genetic Vectors , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/physiopathology , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurturin/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Random Allocation , Rotarod Performance Test
11.
Mol Ther ; 16(10): 1737-44, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18728639

ABSTRACT

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


Subject(s)
Corpus Striatum/metabolism , Dependovirus/genetics , Genetic Vectors , Neurturin/genetics , Transgenes , Animals , Corpus Striatum/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Immunohistochemistry , Macaca fascicularis , Male , Phosphorylation , Tyrosine 3-Monooxygenase/metabolism
12.
Neurobiol Dis ; 27(1): 67-76, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17532642

ABSTRACT

Neurturin (NTN) is a neurotrophic factor with known potential to protect and restore the function of dopaminergic substantia nigra neurons whose degeneration has been most closely linked to the major motor deficits in Parkinson's disease (PD). CERE-120, an adeno-associated virus serotype 2 (AAV2)-based gene delivery vector encoding human NTN, is being developed as a potential therapeutic for PD. In a series of preclinical studies reported herein, CERE-120 delivery to the striatum produced a dose-related neuroprotection of nigrostriatal neurons in the rat 6-hydroxydopamine (6-OHDA) lesion model. Long-lasting efficacy of CERE-120 was evidenced by substantia nigra cell protection, preserved fiber innervation of the striatum, and behavioral recovery for at least 6 months. In addition, striatal infusion of CERE-120 was found to have a safety and tolerability profile devoid of side effects or toxicological responses, for at least 12 months post-treatment, even at dose multiples 125 times that of the lowest efficacious dose tested. These results support the ongoing CERE-120 clinical program in PD patients.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Neurturin/genetics , Parkinsonian Disorders/therapy , Animals , Behavior, Animal , Corpus Striatum/cytology , Corpus Striatum/physiology , Disease Models, Animal , Dopamine/physiology , Female , Genetic Vectors/toxicity , Humans , Male , Nerve Fibers/physiology , Neuroprotective Agents , Oxidopamine , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/physiopathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/cytology , Substantia Nigra/physiology , Sympatholytics
13.
Mov Disord ; 22(8): 1124-32, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17443702

ABSTRACT

Neurturin (NTN) is a potent survival factor for midbrain dopaminergic neurons. CERE-120, an adeno-associated virus type 2 (AAV2) vector encoding human NTN (AAV2-NTN), is currently being developed as a potential therapy for Parkinson's disease. This study examined the bioactivity and safety/tolerability of AAV2-NTN in the aged monkey model of nigrostriatal dopamine insufficiency. Aged rhesus monkeys received unilateral injections of AAV2-NTN into the caudate and putamen, with each animal therefore serving as its own control. Robust expression of NTN within the nigrostriatal system was observed 8 months postadministration. (18)F-fluorodopa imaging using positron emission tomography revealed statistically significant increases in (18)F-fluorodopa uptake in the injected striatum compared with the uninjected side at 4 and 8 months. In addition, at 8 months postadministration, a significant enhancement in tyrosine hydroxylase immunoreactive fibers and an increase in the number of tyrosine hydroxylase immunoreactive cells was observed in the AAV2-NTN injected striatum compared with the uninjected side. Robust activation of phosphorylated extracellular signal-regulated kinase immunoreactivity in the substantia nigra was also observed. Histopathological analyses revealed no adverse effects of AAV2-NTN in the brain. Collectively, these results are consistent with the neurotrophic effects of NTN on the dopaminergic nigrostriatal system and extend the growing body of evidence supporting the concept that AAV2-NTN may have therapeutic benefit for Parkinson's disease.


Subject(s)
Aging/physiology , Corpus Striatum , Dependovirus/metabolism , Disease Models, Animal , Dopamine/metabolism , Neurturin/therapeutic use , Parkinson Disease , Substantia Nigra , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Corpus Striatum/virology , Female , Fluorodeoxyglucose F18/pharmacokinetics , Macaca mulatta , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/virology , Positron-Emission Tomography , Radiopharmaceuticals/pharmacokinetics , Substantia Nigra/metabolism , Substantia Nigra/pathology , Substantia Nigra/virology
14.
Neurobiol Dis ; 26(2): 375-84, 2007 May.
Article in English | MEDLINE | ID: mdl-17336076

ABSTRACT

Huntington's disease (HD) is a devastating neurodegenerative disease characterized by the selective loss of neurons in the striatum and cerebral cortex. This study tested the hypothesis that an adenoassociated viral (AAV2) vector encoding for the trophic factor neurturin (NTN) could provide neuroprotection in the rat 3-nitropropionic acid (3NP) model of HD. Rats received AAV2-NTN (CERE-120), AAV2-eGFP or Vehicle, followed 4 weeks later by the mitochondrial toxin 3NP. 3NP induced motor impairments were observed on the rotarod test, the platform test, and a clinical rating scale in all groups. However, each of these deficits was attenuated by AAV2-NTN (CERE-120). Stereological counts revealed a significant protection of NeuN-ir striatal neurons from 3NP toxicity by AAV2-NTN. These data support the concept that AAV2-NTN might be a valuable treatment for patients with Huntington's disease.


Subject(s)
Corpus Striatum/metabolism , Genetic Therapy/methods , Huntington Disease/therapy , Nerve Degeneration/therapy , Neurons/metabolism , Neurturin/genetics , Animals , Cell Count , Cell Death/genetics , Corpus Striatum/drug effects , Corpus Striatum/physiopathology , Cytoprotection/genetics , DNA-Binding Proteins , Dependovirus/genetics , Disease Models, Animal , Gene Transfer Techniques/trends , Genetic Vectors/genetics , Huntington Disease/chemically induced , Huntington Disease/genetics , Male , Motor Activity/genetics , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/pathology , Neurotoxins , Neurturin/therapeutic use , Nitro Compounds , Nuclear Proteins/metabolism , Propionates , Rats , Rats, Inbred Lew , Recovery of Function/genetics , Treatment Outcome
15.
Mol Ther ; 15(1): 62-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17164776

ABSTRACT

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


Subject(s)
Dependovirus/genetics , Dopamine/metabolism , Genetic Vectors/genetics , Nerve Degeneration/genetics , Neurturin/metabolism , Parkinson Disease/pathology , Parkinson Disease/therapy , Animals , Cell Line , Disease Models, Animal , Dopamine/analogs & derivatives , Gene Expression , Genetic Therapy , Genome, Viral/genetics , Humans , Kinetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/therapy , Neurturin/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Rats , Rats, Sprague-Dawley
16.
Parkinsonism Relat Disord ; 13 Suppl 3: S469-77, 2007.
Article in English | MEDLINE | ID: mdl-18267286

ABSTRACT

PURPOSE: To develop CERE-120 (AAV-NTN) as a novel therapy for Parkinson's disease (PD) that might restore function of degenerating dopamine neurons and prevent further degeneration. SCOPE: A nonclinical program demonstrated that NTN expression can be predictably controlled following CERE-120 administration, provides clear evidence of efficacy in numerous animal models and is safe at dose multiples that far exceed those required for efficacy. Preliminary, open label evidence in PD subjects offers corroborative support for these observations. CONCLUSIONS: CERE-120 may represent an important, novel therapy for PD, though the clinical data require confirmation with additional clinical tests, including an ongoing multi-center, double-blinded controlled trial.


Subject(s)
Genetic Therapy/methods , Neurturin/physiology , Parkinson Disease/therapy , Animals , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Neuropeptides , Neurturin/genetics
17.
Ann Neurol ; 60(6): 706-15, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17192932

ABSTRACT

OBJECTIVE: We tested the hypothesis that gene delivery of the trophic factor neurturin could preserve motor function and protect nigrostriatal circuitry in hemiparkinsonian monkeys. METHODS: An adeno-associated virus-based vector encoding human neurturin (AAV2-NTN; also called CERE-120) was injected into the striatum and substantia nigra of monkeys 4 days after a unilateral intracarotid injection of N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) rendered them hemiparkinsonian. Control hemiparkinsonian monkeys received either AAV2 encoding green fluorescent protein or formulation buffer. RESULTS: Although stable deficits were seen in all control monkeys, AAV2-NTN significantly improved MPTP-induced motor impairments by 80 to 90% starting at approximately month 4 and lasting until the end of the experiment (month 10). AAV2-NTN significantly preserved nigral neurons, significantly preserved striatal dopaminergic innervation, and activated phospho-extracellular signal-regulated kinase, consistent with a mechanism involving a trophic factor-initiated molecular cascade. Histological analyses of numerous brain regions, including the cerebellum, showed normal cytoarchitecture and no aberrant pathology. INTERPRETATION: These data demonstrate that AAV2-NTN (CERE-120) can preserve function and anatomy in degenerating nigrostriatal neurons and are supportive of ongoing clinical tests in Parkinson's disease patients.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/therapeutic use , Neurturin/genetics , Parkinsonian Disorders/therapy , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Cytoprotection/genetics , Disease Models, Animal , Dopamine/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Transfer Techniques , Genetic Vectors/genetics , Genetic Vectors/metabolism , Macaca mulatta , Male , Nerve Regeneration/genetics , Neural Pathways/metabolism , Neural Pathways/pathology , Neural Pathways/physiopathology , Neurons/metabolism , Neurons/pathology , Neurturin/metabolism , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , Recovery of Function/genetics , Substantia Nigra/metabolism , Substantia Nigra/pathology , Substantia Nigra/physiopathology , Treatment Outcome
18.
Proc Natl Acad Sci U S A ; 103(24): 9345-50, 2006 Jun 13.
Article in English | MEDLINE | ID: mdl-16751280

ABSTRACT

Huntington's disease (HD) is a fatal, genetic, neurological disorder resulting from a trinucleotide repeat expansion in the gene that encodes for the protein huntingtin. These excessive repeats confer a toxic gain of function on huntingtin, which leads to the degeneration of striatal and cortical neurons and a devastating motor, cognitive, and psychological disorder. Trophic factor administration has emerged as a compelling potential therapy for a variety of neurodegenerative disorders, including HD. We previously demonstrated that viral delivery of glial cell line-derived neurotrophic factor (GDNF) provides structural and functional neuroprotection in a rat neurotoxin model of HD. In this report we demonstrate that viral delivery of GDNF into the striatum of presymptomatic mice ameliorates behavioral deficits on the accelerating rotorod and hind limb clasping tests in transgenic HD mice. Behavioral neuroprotection was associated with anatomical preservation of the number and size of striatal neurons from cell death and cell atrophy. Additionally, GDNF-treated mice had a lower percentage of neurons containing mutant huntingtin-stained inclusion bodies, a hallmark of HD pathology. These data further support the concept that viral vector delivery of GDNF may be a viable treatment for patients suffering from HD.


Subject(s)
Gene Transfer Techniques , Glial Cell Line-Derived Neurotrophic Factors/metabolism , Huntington Disease/pathology , Huntington Disease/physiopathology , Neuroprotective Agents/metabolism , Animals , Behavior, Animal/physiology , Cell Death , Corpus Striatum/cytology , Corpus Striatum/metabolism , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Female , Genetic Therapy , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Glial Cell Line-Derived Neurotrophic Factors/genetics , Glial Cell Line-Derived Neurotrophic Factors/therapeutic use , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/therapy , Inclusion Bodies/chemistry , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Nuclear Proteins/metabolism , Rats
19.
Neurobiol Aging ; 24(6): 861-3, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12927768

ABSTRACT

Aging-related differences in the ability of cortical cholinergic inputs to respond to local depolarization was assessed in young (3-6 months) and old (26-33 months) awake rats using in vivo microdialysis in the absence of an inhibitor of acetylcholinesterase. Rats were perfused, using a within-subjects, repeated session design, with vehicle (aCSF) or K(+) (25, 50, 100 mM). Perfusion of K(+) resulted in a dose-dependent increase in cortical ACh efflux with comparable efflux seen between the two ages following 25 mM (50%) and 50 mM (100%) K(+). In contrast, aged rats exhibited a marked attenuation (330%) in ACh efflux relative to young adult rats (650%). These data reveal aging-related decreases in the responsiveness of cortical cholinergic afferents, tested under physiologically relevant conditions, to local depolarization and may provide a neuronal mechanism contributing to the cognitive deficits reported in normal aging- and age-related pathological conditions.


Subject(s)
Acetylcholine/metabolism , Aging/metabolism , Cholinergic Fibers/metabolism , Neurons/metabolism , Prefrontal Cortex/metabolism , Acetylcholinesterase/metabolism , Afferent Pathways/metabolism , Age Factors , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Microdialysis/methods , Neurons/drug effects , Potassium/pharmacology , Prefrontal Cortex/drug effects , Rats , Rats, Inbred F344
20.
Neurobiol Aging ; 23(3): 467-77, 2002.
Article in English | MEDLINE | ID: mdl-11959409

ABSTRACT

Pre-existing trauma to basal forebrain corticopetal cholinergic neurons has been hypothesized to render this system vulnerable to age-related processes. The present longitudinal study assessed the interactions between the effects of partial cortical cholinergic deafferentation and aging on sustained attention performance. After pre-surgical training, animals were given sham-surgery or bilateral infusions of the immunotoxin 192 IgG-saporin into the basal forebrain. The lesion was intended to yield a limited loss of cortical cholinergic inputs and thus to produce minor immediate effects on sustained attention performance. All animals were tested continuously until age 36 months. The attentional performance of lesioned and sham-lesioned animals did not dissociate until age 31 months, when the lesioned animals exhibited an impairment in overall sustained attention performance. Importantly, this impairment interacted with the effects of time-on-task, and thus reflected a specific impairment in attentional processes. These results support the notion that pre-existing damage to the basal forebrain corticopetal cholinergic neurons yields age-related impairments in the attentional capabilities that depend on the integrity of this neuronal system.


Subject(s)
Aging/physiology , Attention/physiology , Cerebral Cortex/physiology , Cholinergic Fibers/physiology , Afferent Pathways/pathology , Afferent Pathways/physiopathology , Aging/pathology , Animals , Behavior, Animal/physiology , Body Weight/physiology , Cerebral Cortex/pathology , Cholinergic Fibers/pathology , Conditioning, Operant/physiology , Male , Rats , Rats, Inbred BN , Rats, Inbred F344 , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...