Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Mol Ther Methods Clin Dev ; 26: 384-393, 2022 Sep 08.
Article in English | MEDLINE | ID: mdl-36034772

ABSTRACT

Multiple studies have examined the transduction characteristics of different AAV serotypes in the mouse brain, where they can exhibit significantly different patterns of transduction. The pattern of transduction also varies with the route of administration. Much less information exists for the transduction characteristics in large-brained animals. Large animal models have brains that are closer in size and organization to the human brain, such as being gyrencephalic compared to the lissencephalic rodent brains, pathway organization, and certain electrophysiologic properties. Large animal models are used as translational intermediates to develop gene therapies to treat human diseases. Various AAV serotypes and routes of delivery have been used to study the correction of pathology in the brain in lysosomal storage diseases. In this study, we evaluated the ability of selected AAV serotypes to transduce cells in the cat brain when delivered into the cerebrospinal fluid via the cisterna magna. We previously showed that AAV1 transduced significantly greater numbers of cells than AAV9 in the cat brain by this route. In the present study, we evaluated serotypes closely related to AAVs 1 and 9 (AAVs 6, AS, hu32) that may mediate more extensive transduction, as well as AAVs 4 and 5, which primarily transduce choroid plexus epithelial (CPE) and ependymal lining cells in the rodent brain. The related serotypes tended to have similar patterns of transduction but were divergent in some specific brain structures.

2.
PLoS One ; 16(12): e0260805, 2021.
Article in English | MEDLINE | ID: mdl-34855883

ABSTRACT

BACKGROUND: Hereditary angioedema (HAE) is a rare disease that manifests as recurrent and debilitating angioedema attacks, significantly impacting patients' quality of life. OBJECTIVE: To assess communication dynamics between patients with HAE and treating physicians and the impact this has on the treatment of HAE in the United States. METHODS: This observational study used an institutional review board-approved protocol to collect four sources of patient-physician communication data from the period between January 2015 and May 2017: in-office conversations between patients aged ≥18 years with HAE and physicians, follow-up dictations with physicians, telephone interviews with patients and physicians, and publicly available social media posts from patients. Participant language was qualitatively assessed and key communication elements and communication gaps identified. RESULTS: Twenty-five in-office conversations, 14 follow-up physician dictations, and 17 telephone interviews were conducted with a total of 29 unique patients, 4 caregivers, and 14 physicians. In-office conversations were generally physician-driven and focused primarily on symptom frequency, location, and severity; lexicon from both parties centered on "episodes" and "swelling." During visits, impact on quality of life was not routinely assessed by physicians nor discussed proactively by patients; however, during telephone interviews and online, patients frequently described the multifaceted burden of HAE. Patients highlighted the difficulties they experience by using repetition, emphasis, and metaphors; they also varied the descriptors used for attacks depending on the communication goal. Physicians used intensifiers to emphasize the necessity of rescue medication access, whereas prophylactic treatments were positioned as an option for frequent or laryngeal attacks. CONCLUSION: Vocabulary differences suggest that the full impact of HAE is not consistently communicated by patients to physicians during clinical visits, indicating the potential for misaligned understanding of disease burden. A patient-driven, rather than physician-driven approach to the discussions may elicit valuable information that could help to optimize treatment approaches.


Subject(s)
Angioedemas, Hereditary/epidemiology , Communication , Patient Reported Outcome Measures , Physicians/psychology , Quality of Life , Severity of Illness Index , Adolescent , Adult , Aged , Angioedemas, Hereditary/physiopathology , Female , Humans , Male , Middle Aged , Surveys and Questionnaires , United States , Young Adult
3.
Mol Genet Metab ; 133(4): 378-385, 2021 08.
Article in English | MEDLINE | ID: mdl-34154922

ABSTRACT

Mucopolysaccharidosis (MPS) VII is a lysosomal storage disorder characterized by deficient ß-glucuronidase activity, leading to accumulation of incompletely degraded heparan, dermatan and chondroitin sulfate glycosaminoglycans. Patients with MPS VII exhibit progressive spinal deformity, which decreases quality of life. Previously, we demonstrated that MPS VII dogs exhibit impaired initiation of secondary ossification in the vertebrae and long bones. The objective of this study was to build on these findings and comprehensively characterize how vertebral bone disease manifests progressively in MPS VII dogs throughout postnatal growth. Vertebrae were collected postmortem from MPS VII and healthy control dogs at seven ages ranging from 9 to 365 days. Microcomputed tomography and histology were used to characterize bone properties in primary and secondary ossification centers. Serum was analyzed for bone turnover biomarkers. Results demonstrated that not only was secondary ossification delayed in MPS VII vertebrae, but that it progressed aberrantly and was markedly diminished even at 365 days-of-age. Within primary ossification centers, bone volume fraction and bone mineral density were significantly lower in MPS VII at 180 and 365 days-of-age. MPS VII growth plates exhibited significantly lower proliferative and hypertrophic zone cellularity at 90 days-of-age, while serum bone-specific alkaline phosphatase (BAP) was significantly lower in MPS VII dogs at 180 days-of-age. Overall, these findings establish that vertebral bone formation is significantly diminished in MPS VII dogs in both primary and secondary ossification centers during postnatal growth.


Subject(s)
Bone Diseases/physiopathology , Disease Progression , Mucopolysaccharidosis VII/complications , Spine/pathology , Animals , Animals, Newborn , Bone Diseases/genetics , Bone and Bones/pathology , Dogs , Female , Growth and Development , Male , Mucopolysaccharidosis VII/genetics , Osteogenesis
4.
Mol Ther Methods Clin Dev ; 20: 247-257, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33473358

ABSTRACT

Adeno-associated viral (AAV) vectors have emerged as the preferred platform for in vivo gene transfer because of their combined efficacy and safety. However, insertional mutagenesis with the subsequent development of hepatocellular carcinomas (HCCs) has been recurrently noted in newborn mice treated with high doses of AAV, and more recently, the association of wild-type AAV integrations in a subset of human HCCs has been documented. Here, we address, in a comprehensive, prospective study, the long-term risk of tumorigenicity in young adult mice following delivery of single-stranded AAVs targeting liver. HCC incidence in mice treated with therapeutic and reporter AAVs was low, in contrast to what has been previously documented in mice treated as newborns with higher doses of AAV. Specifically, HCCs developed in 6 out 76 of AAV-treated mice, and a pathogenic integration of AAV was found in only one tumor. Also, no evidence of liver tumorigenesis was found in juvenile AAV-treated mucopolysaccharidosis type VI (MPS VI) cats followed as long as 8 years after vector administration. Together, our results support the low risk of tumorigenesis associated with AAV-mediated gene transfer targeting juvenile/young adult livers, although constant monitoring of subjects enrolled in AAV clinical trial is advisable.

5.
Brain ; 143(7): 2058-2072, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32671406

ABSTRACT

Intravascular injection of certain adeno-associated virus vector serotypes can cross the blood-brain barrier to deliver a gene into the CNS. However, gene distribution has been much more limited within the brains of large animals compared to rodents, rendering this approach suboptimal for treatment of the global brain lesions present in most human neurogenetic diseases. The most commonly used serotype in animal and human studies is 9, which also has the property of being transported via axonal pathways to distal neurons. A small number of other serotypes share this property, three of which were tested intravenously in mice compared to 9. Serotype hu.11 transduced fewer cells in the brain than 9, rh8 was similar to 9, but hu.32 mediated substantially greater transduction than the others throughout the mouse brain. To evaluate the potential for therapeutic application of the hu.32 serotype in a gyrencephalic brain of larger mammals, a hu.32 vector expressing the green fluorescent protein reporter gene was evaluated in the cat. Transduction was widely distributed in the cat brain, including in the cerebral cortex, an important target since mental retardation is an important component of many of the human neurogenetic diseases. The therapeutic potential of a hu.32 serotype vector was evaluated in the cat homologue of the human lysosomal storage disease alpha-mannosidosis, which has globally distributed lysosomal storage lesions in the brain. Treated alpha-mannosidosis cats had reduced severity of neurological signs and extended life spans compared to untreated cats. The extent of therapy was dose dependent and intra-arterial injection was more effective than intravenous delivery. Pre-mortem, non-invasive magnetic resonance spectroscopy and diffusion tensor imaging detected differences between the low and high doses, and showed normalization of grey and white matter imaging parameters at the higher dose. The imaging analysis was corroborated by post-mortem histological analysis, which showed reversal of histopathology throughout the brain with the high dose, intra-arterial treatment. The hu.32 serotype would appear to provide a significant advantage for effective treatment of the gyrencephalic brain by systemic adeno-associated virus delivery in human neurological diseases with widespread brain lesions.


Subject(s)
Brain/virology , Dependovirus , Disease Models, Animal , Genetic Therapy/methods , Genetic Vectors , alpha-Mannosidosis/genetics , Animals , Brain/pathology , Cats , Gene Transfer Techniques , Transduction, Genetic
6.
Mol Ther ; 28(6): 1455-1463, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32330426

ABSTRACT

Mucopolysaccharidosis type I (MPS I) is an autosomal recessive lysosomal storage disease characterized by severe phenotypes, including corneal clouding. MPS I is caused by mutations in alpha-l-iduronidase (IDUA), a ubiquitous enzyme that catalyzes the hydrolysis of glycosaminoglycans. Currently, no treatment exists to address MPS I corneal clouding other than corneal transplantation, which is complicated by a high risk for rejection. Investigation of an adeno-associated virus (AAV) IDUA gene addition strategy targeting the corneal stroma addresses this deficiency. In MPS I canines with early or advanced corneal disease, a single intrastromal AAV8G9-IDUA injection was well tolerated at all administered doses. The eyes with advanced disease demonstrated resolution of corneal clouding as early as 1 week post-injection, followed by sustained corneal transparency until the experimental endpoint of 25 weeks. AAV8G9-IDUA injection in the MPS I canine eye with early corneal disease prevented the development of advanced corneal changes while restoring clarity. Biodistribution studies demonstrated vector genomes in ocular compartments other than the cornea and in some systemic organs; however, a capsid antibody response was detected in only the highest dosed subject. Collectively, the results suggest that intrastromal AAV8G9-IDUA therapy prevents and reverses visual impairment associated with MPS I corneal clouding.


Subject(s)
Corneal Diseases/etiology , Corneal Diseases/therapy , Gene Transfer Techniques , Genetic Therapy , Mucopolysaccharidosis I/complications , Mucopolysaccharidosis I/genetics , Animals , Animals, Genetically Modified , Corneal Diseases/diagnosis , Dependovirus/genetics , Disease Models, Animal , Dogs , Female , Fluorescent Antibody Technique , Gene Expression , Gene Knockdown Techniques , Genes, Reporter , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Iduronidase/genetics , Male , Transgenes , Treatment Outcome
7.
J Feline Med Surg ; 21(10): 882-886, 2019 10.
Article in English | MEDLINE | ID: mdl-30339054

ABSTRACT

OBJECTIVES: The aim of this study was to determine the pharmacokinetics of furosemide in cats following intravenous (IV), oral and transdermal administration. METHODS: This study used six healthy adult cats in a three-phase design to compare plasma furosemide concentrations in cats that received one IV 2 mg/kg dose of furosemide, one oral 2 mg/kg dose of furosemide and 3 days of q12h dosing with 2 mg/kg furosemide transdermally applied to the ear pinna. RESULTS: After IV administration the elimination half-life was (mean and coefficient of variation) 2.25 h (72%), systemic clearance was 149 ml/kg/h (27.4%) and volume of distribution was 227 ml/kg (22%). After oral administration the terminal half-life was 1.2 h (18.7%), peak concentration was 3.4 µg/ml (51.7%) and bioavailability was 48.4%. The transdermal plasma concentrations were undetectable or very low at most time points, and pharmacokinetics were not determined from the transdermal dose. CONCLUSIONS AND RELEVANCE: Furosemide was rapidly eliminated in cats after oral and IV administration and is probably best administered orally at least q12h in cats with heart failure. The oral dose absorbed was approximately 50%, but the absorption from transdermal administration was negligible.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Cats/metabolism , Furosemide/pharmacokinetics , Administration, Cutaneous , Administration, Oral , Administration, Topical , Analgesics, Opioid/administration & dosage , Animals , Biological Availability , Female , Furosemide/administration & dosage , Half-Life , Infusions, Intravenous/veterinary , Injections, Intravenous/veterinary , Male
8.
Blood Adv ; 2(9): 987-999, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29720491

ABSTRACT

Hematopoietic stem-cell gene therapy is a promising treatment of X-linked severe combined immunodeficiency disease (SCID-X1), but currently, it requires recipient conditioning, extensive cell manipulation, and sophisticated facilities. With these limitations in mind, we explored a simpler therapeutic approach to SCID-X1 treatment by direct IV administration of foamy virus (FV) vectors in the canine model. FV vectors were used because they have a favorable integration site profile and are resistant to serum inactivation. Here, we show improved efficacy of our in vivo gene therapy platform by mobilization with granulocyte colony-stimulating factor (G-CSF) and AMD3100 before injection of an optimized FV vector incorporating the human phosphoglycerate kinase enhancerless promoter. G-CSF/AMD3100 mobilization before FV vector delivery accelerated kinetics of CD3+ lymphocyte recovery, promoted thymopoiesis, and increased immune clonal diversity. Gene-corrected T lymphocytes exhibited a normal CD4:CD8 ratio and a broad T-cell receptor repertoire and showed restored γC-dependent signaling function. Treated animals showed normal primary and secondary antibody responses to bacteriophage immunization and evidence for immunoglobulin class switching. These results demonstrate safety and efficacy of an accessible, portable, and translatable platform with no conditioning regimen for the treatment of SCID-X1 and other genetic diseases.


Subject(s)
Dog Diseases , Genetic Therapy , Genetic Vectors/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Heterocyclic Compounds/pharmacology , Spumavirus , X-Linked Combined Immunodeficiency Diseases , Animals , Benzylamines , CD4-CD8 Ratio , Cyclams , Disease Models, Animal , Dog Diseases/blood , Dog Diseases/genetics , Dog Diseases/therapy , Dogs , Humans , Phosphoglycerate Kinase/genetics , X-Linked Combined Immunodeficiency Diseases/blood , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/therapy , X-Linked Combined Immunodeficiency Diseases/veterinary
9.
Hum Gene Ther ; 29(1): 15-24, 2018 01.
Article in English | MEDLINE | ID: mdl-28806897

ABSTRACT

Delivery of adeno-associated viral (AAV) vectors into the cerebrospinal fluid (CSF) can achieve gene transfer to cells throughout the brain and spinal cord, potentially making many neurological diseases tractable gene therapy targets. Identifying the optimal route of CSF access for intrathecal AAV delivery will be a critical step in translating this approach to clinical practice. We previously demonstrated that vector injection into the cisterna magna is a safe and effective method for intrathecal AAV delivery in nonhuman primates; however, this procedure is not commonly used in clinical practice. More routine methods of administration into the CSF are now being explored, including intracerebroventricular (ICV) injection and injection through a lumbar puncture. In this study, we compared ICV and intracisternal (IC) AAV administration in dogs. We also evaluated vector administration via lumbar puncture in nonhuman primates, with some animals placed in the Trendelenburg position after injection, a maneuver that has been suggested to improve cranial distribution of vector. In the dog study, ICV and IC vector administration resulted in similarly efficient transduction throughout the brain and spinal cord. However, animals in the ICV cohort developed encephalitis associated with a T-cell response to the transgene product, a phenomenon that was not observed in the IC cohort. In the nonhuman primate study, transduction efficiency was not improved by placing animals in the Trendelenburg position after injection. These findings illustrate important limitations of commonly used methods for CSF access in the context of AAV delivery, and will be important for informing the selection of a route of administration for first-in-human studies.


Subject(s)
Central Nervous System Diseases/therapy , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Animals , Central Nervous System Diseases/genetics , Dependovirus/genetics , Dogs , Genetic Therapy/methods , Genetic Vectors/cerebrospinal fluid , Haplorhini , Head-Down Tilt , Infusions, Intraventricular , Injections, Spinal , Models, Animal , Spinal Puncture
10.
J Neurosci Res ; 94(11): 1007-17, 2016 11.
Article in English | MEDLINE | ID: mdl-27638585

ABSTRACT

Globoid cell leukodystrophy (GLD), or Krabbe's disease, is a debilitating and always fatal pediatric neurodegenerative disease caused by a mutation in the gene encoding the hydrolytic enzyme galactosylceramidase (GALC). In the absence of GALC, progressive loss of myelin and accumulation of a neurotoxic substrate lead to incapacitating loss of motor and cognitive function and death, typically by 2 years of age. Currently, there is no cure. Recent convincing evidence of the therapeutic potential of combining gene and cell therapies in the murine model of GLD has accelerated the requirement for validated markers of disease to evaluate therapeutic efficacy. Here we demonstrate clinically relevant and quantifiable measures of central (CNS) and peripheral (PNS) nervous system disease progression in the naturally occurring canine model of GLD. As measured by brainstem auditory-evoked response testing, GLD dogs demonstrated a significant increase in I-V interpeak latency and hearing threshold at all time points. Motor nerve conduction velocities (NCVs) in GLD dogs were significantly lower than normal by 12-16 weeks of age, and sensory NCV was significantly lower than normal by 8-12 weeks of age, serving as a sensitive indicator of peripheral nerve dysfunction. Post-mortem histological evaluations confirmed neuroimaging and electrodiagnostic assessments and detailed loss of myelin and accumulation of storage product in the CNS and the PNS. Additionally, cerebrospinal fluid psychosine concentrations were significantly elevated in GLD dogs, demonstrating potential as a biochemical marker of disease. These data demonstrate that CNS and PNS disease progression can be quantified over time in the canine model of GLD with tools identical to those used to assess human patients. © 2016 Wiley Periodicals, Inc.


Subject(s)
Evoked Potentials, Auditory, Brain Stem/genetics , Leukodystrophy, Globoid Cell/complications , Leukodystrophy, Globoid Cell/genetics , Nervous System Diseases , Animals , Disease Models, Animal , Dogs , Electric Stimulation , Female , Galactosylceramidase/genetics , Humans , Leukodystrophy, Globoid Cell/diagnostic imaging , Leukodystrophy, Globoid Cell/veterinary , Magnetic Resonance Imaging , Male , Mutation, Missense/genetics , Nervous System/diagnostic imaging , Nervous System/pathology , Nervous System/physiopathology , Nervous System Diseases/diagnostic imaging , Nervous System Diseases/etiology , Nervous System Diseases/metabolism , Nervous System Diseases/therapy , Neural Conduction/genetics , Psychosine/cerebrospinal fluid
11.
Mol Genet Metab ; 119(1-2): 124-30, 2016 09.
Article in English | MEDLINE | ID: mdl-27386755

ABSTRACT

High fidelity animal models of human disease are essential for preclinical evaluation of novel gene and protein therapeutics. However, these studies can be complicated by exaggerated immune responses against the human transgene. Here we demonstrate that dogs with a genetic deficiency of the enzyme α-l-iduronidase (IDUA), a model of the lysosomal storage disease mucopolysaccharidosis type I (MPS I), can be rendered immunologically tolerant to human IDUA through neonatal exposure to the enzyme. Using MPS I dogs tolerized to human IDUA as neonates, we evaluated intrathecal delivery of an adeno-associated virus serotype 9 vector expressing human IDUA as a therapy for the central nervous system manifestations of MPS I. These studies established the efficacy of the human vector in the canine model, and allowed for estimation of the minimum effective dose, providing key information for the design of first-in-human trials. This approach can facilitate evaluation of human therapeutics in relevant animal models, and may also have clinical applications for the prevention of immune responses to gene and protein replacement therapies.


Subject(s)
Enzyme Replacement Therapy , Iduronidase/genetics , Lysosomal Storage Diseases/therapy , Mucopolysaccharidosis I/therapy , Animals , Disease Models, Animal , Dogs , Genetic Therapy , Genetic Vectors , Glycosaminoglycans/metabolism , Humans , Iduronidase/deficiency , Iduronidase/therapeutic use , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/pathology , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/pathology , Transgenes
12.
J Pharmacol Exp Ther ; 358(2): 254-61, 2016 08.
Article in English | MEDLINE | ID: mdl-27307499

ABSTRACT

Niemann-Pick type C (NPC) 1 disease is a rare, inherited, neurodegenerative disease. Clear evidence of the therapeutic efficacy of 2-hydroxypropyl-ß-cyclodextrin (HPßCD) in animal models resulted in the initiation of a phase I/IIa clinical trial in 2013 and a phase IIb/III trial in 2015. With clinical trials ongoing, validation of a biomarker to track disease progression and serve as a supporting outcome measure of therapeutic efficacy has become compulsory. In this study, we evaluated calcium-binding protein calbindin D-28K (calbindin) concentrations in the cerebrospinal fluid (CSF) as a biomarker of NPC1 disease. In the naturally occurring feline model, CSF calbindin was significantly elevated at 3 weeks of age, prior to the onset of cerebellar dysfunction, and steadily increased to >10-fold over normal at end-stage disease. Biweekly intrathecal administration of HPßCD initiated prior to the onset of neurologic dysfunction completely normalized CSF calbindin in NPC1 cats at all time points analyzed when followed up to 78 weeks of age. Initiation of HPßCD after the onset of clinical signs (16 weeks of age) resulted in a delayed reduction of calbindin levels in the CSF. Evaluation of CSF from patients with NPC1 revealed that calbindin concentrations were significantly elevated compared with CSF samples collected from unaffected patients. Off-label treatment of patients with NPC1 with miglustat, an inhibitor of glycosphingolipid biosynthesis, significantly decreased CSF calbindin compared with pretreatment concentrations. These data suggest that the CSF calbindin concentration is a sensitive biomarker of NPC1 disease that could be instrumental as an outcome measure of therapeutic efficacy in ongoing clinical trials.


Subject(s)
Biomarkers/cerebrospinal fluid , Calbindin 1/cerebrospinal fluid , Disease Progression , Niemann-Pick Disease, Type C/cerebrospinal fluid , 2-Hydroxypropyl-beta-cyclodextrin , Adolescent , Adult , Animals , Cats , Child , Child, Preschool , Female , Glycosphingolipids/biosynthesis , Humans , Infant , Male , Middle Aged , Niemann-Pick Disease, Type C/metabolism , Time Factors , Young Adult , beta-Cyclodextrins/pharmacology
13.
J Neuropathol Exp Neurol ; 75(1): 35-43, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26671987

ABSTRACT

α-Mannosidosis (AMD) is an autosomal recessively inherited lysosomal storage disorder affecting brain function and structure. We performed ex vivo and in vivo diffusion tensor imaging (DTI) on the brains of AMD-affected cats to assess gray and white matter abnormalities. A multi-atlas approach was used to generate a brain template to process the ex vivo DTI data. The probabilistic label method was used to measure fractional anisotropy (FA), mean diffusivity, axial diffusivity, and radial diffusivity values from gray and white matter regions from ex vivo DTI. Regional analysis from various regions of the gray matter (frontal cortex, cingulate gyrus, caudate nucleus, hippocampus, thalamus, and occipital cortex), and white matter (corpus callosum, corticospinal tract, cerebral peduncle, external and internal capsule) was also performed on both ex vivo and in vivo DTI. Ex vivo DTI revealed significantly reduced FA from both gray and white matter regions in AMD-affected cats compared to controls. Significantly reduced FA was also observed from in vivo DTI of AMD-affected cats compared to controls, with lower FA values observed in all white matter regions. We also observed significantly increased axial and radial diffusivity values in various gray and white matter regions in AMD cats from both ex vivo and in vivo DTI data. Imaging findings were correlated with histopathologic analyses suggesting that DTI studies can further aid in the characterization of AMD by assessing the microstructural abnormalities in both white and gray matter.


Subject(s)
Diffusion Tensor Imaging/methods , Gray Matter/metabolism , Gray Matter/pathology , White Matter/metabolism , White Matter/pathology , alpha-Mannosidosis/metabolism , Animals , Animals, Genetically Modified , Cats , alpha-Mannosidosis/diagnosis
14.
Mol Ther ; 24(2): 206-216, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26447927

ABSTRACT

Mucopolysaccharidosis VII (MPS VII) is a lysosomal storage disease arising from mutations in ß-d-glucuronidase (GUSB), which results in glycosaminoglycan (GAG) accumulation and a variety of clinical manifestations including neurological disease. Herein, MPS VII dogs were injected intravenously (i.v.) and/or intrathecally (i.t.) via the cisterna magna with AAV9 or AAVrh10 vectors carrying the canine GUSB cDNA. Although i.v. injection alone at 3 days of age resulted in normal cerebrospinal fluid (CSF) GUSB activity, brain tissue homogenates had only ~1 to 6% normal GUSB activity and continued to have elevated GAG storage. In contrast, i.t. injection at 3 weeks of age resulted in CSF GUSB activity 44-fold normal while brain tissue homogenates had >100% normal GUSB activity and reduced GAGs compared with untreated dogs. Markers for secondary storage and inflammation were eliminated in i.t.-treated dogs and reduced in i.v.-treated dogs compared with untreated dogs. Given that i.t.-treated dogs expressed higher levels of GUSB in the CNS tissues compared to those treated i.v., we conclude that i.t. injection of AAV9 or AAVrh10 vectors is more effective than i.v. injection alone in the large animal model of MPS VII.


Subject(s)
Central Nervous System Diseases/therapy , Genetic Therapy/methods , Glucuronidase/genetics , Mucopolysaccharidosis VII/therapy , Animals , Animals, Newborn , Central Nervous System Diseases/genetics , Central Nervous System Diseases/metabolism , Dependovirus/genetics , Disease Models, Animal , Dogs , Genetic Vectors/administration & dosage , Glucuronidase/cerebrospinal fluid , Glycosaminoglycans/metabolism , Injections, Intravenous , Injections, Spinal , Male , Mucopolysaccharidosis VII/complications , Mucopolysaccharidosis VII/genetics , Mucopolysaccharidosis VII/metabolism
15.
Mol Ther ; 24(1): 26-33, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26354342

ABSTRACT

Lysosomal storage diseases (LSDs) are debilitating neurometabolic disorders for most of which long-term effective therapies have not been developed. Gene therapy is a potential treatment but a critical barrier to treating the brain is the need for global correction. We tested the efficacy of cisterna magna infusion of adeno-associated virus type 1 (AAV1) expressing feline alpha-mannosidase gene in the postsymptomatic alpha-mannosidosis (AMD) cat, a homologue of the human disease. Lysosomal alpha-mannosidase (MANB) activity in the cerebrospinal fluid (CSF) and serum were increased above the control values in untreated AMD cats. Clinical neurological signs were delayed in onset and reduced in severity. The lifespan of the treated cats was significantly extended. Postmortem histopathology showed resolution of lysosomal storage lesions throughout the brain. MANB activity in brain tissue was significantly above the levels of untreated tissues. The results demonstrate that a single cisterna magna injection of AAV1 into the CSF can mediate widespread neuronal transduction of the brain and meaningful clinical improvement. Thus, cisterna magna gene delivery by AAV1 appears to be a viable strategy for treatment of the whole brain in AMD and should be applicable to many of the neurotropic LSDs as well as other neurogenetic disorders.


Subject(s)
Cat Diseases/therapy , Cisterna Magna/metabolism , Dependovirus/genetics , alpha-Mannosidase/genetics , alpha-Mannosidosis/veterinary , Age of Onset , Animals , Brain/enzymology , Cat Diseases/pathology , Cats , Disease Models, Animal , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Injections , Lysosomes/metabolism , alpha-Mannosidase/blood , alpha-Mannosidase/cerebrospinal fluid , alpha-Mannosidase/metabolism , alpha-Mannosidosis/pathology , alpha-Mannosidosis/therapy
16.
Mol Ther ; 23(8): 1298-1307, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26022732

ABSTRACT

The potential host immune response to a nonself protein poses a fundamental challenge for gene therapies targeting recessive diseases. We demonstrate in both dogs and nonhuman primates that liver-directed gene transfer using an adeno-associated virus (AAV) vector in neonates induces a persistent state of immunological tolerance to the transgene product, substantially improving the efficacy of subsequent vector administration targeting the central nervous system (CNS). We applied this approach to a canine model of mucopolysaccharidosis type I (MPS I), a progressive neuropathic lysosomal storage disease caused by deficient activity of the enzyme α-l-iduronidase (IDUA). MPS I dogs treated systemically in the first week of life with a vector expressing canine IDUA did not develop antibodies against the enzyme and exhibited robust expression in the CNS upon intrathecal AAV delivery at 1 month of age, resulting in complete correction of brain storage lesions. Newborn rhesus monkeys treated systemically with AAV vector expressing human IDUA developed tolerance to the transgene, resulting in high cerebrospinal fluid (CSF) IDUA expression and no antibody induction after subsequent CNS gene therapy. These findings suggest that inducing tolerance to the transgene product during a critical period in immunological development can improve the efficacy and safety of gene therapy.


Subject(s)
Central Nervous System/metabolism , Dependovirus/genetics , Genetic Therapy/methods , Iduronidase/genetics , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/therapy , Animals , Animals, Newborn , Disease Models, Animal , Dogs , Female , Gene Transfer Techniques , Genetic Vectors , HEK293 Cells , Humans , Iduronidase/deficiency , Macaca mulatta , Transgenes
17.
Sci Transl Med ; 7(276): 276ra26, 2015 Feb 25.
Article in English | MEDLINE | ID: mdl-25717099

ABSTRACT

Niemann-Pick type C1 (NPC) disease is a lysosomal storage disease caused by mutations in the NPC1 gene, leading to an increase in unesterified cholesterol and several sphingolipids, and resulting in hepatic disease and progressive neurological disease. We show that subcutaneous administration of the pharmaceutical excipient 2-hydroxypropyl-ß-cyclodextrin (HPßCD) to cats with NPC disease ameliorated hepatic disease, but doses sufficient to reduce neurological disease resulted in pulmonary toxicity. However, direct administration of HPßCD into the cisterna magna of presymptomatic cats with NPC disease prevented the onset of cerebellar dysfunction for greater than a year and resulted in a reduction in Purkinje cell loss and near-normal concentrations of cholesterol and sphingolipids. Moreover, administration of intracisternal HPßCD to NPC cats with ongoing cerebellar dysfunction slowed disease progression, increased survival time, and decreased the accumulation of brain gangliosides. An increase in hearing threshold was identified as a potential adverse effect. These studies in a feline animal model have provided critical data on efficacy and safety of drug administration directly into the central nervous system that will be important for advancing HPßCD into clinical trials.


Subject(s)
Cisterna Magna/pathology , Cisterna Magna/physiopathology , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/physiopathology , Purkinje Cells/pathology , beta-Cyclodextrins/therapeutic use , 2-Hydroxypropyl-beta-cyclodextrin , Aging/pathology , Alanine Transaminase/blood , Animals , Ataxia/blood , Ataxia/complications , Ataxia/pathology , Auditory Threshold , Calbindins/metabolism , Cats , Cell Death , Fluorescent Antibody Technique , G(M2) Ganglioside/metabolism , Inflammation/complications , Inflammation/pathology , Injections, Subcutaneous , Liver/pathology , Liver Diseases/blood , Liver Diseases/complications , Liver Diseases/pathology , Lung/pathology , Niemann-Pick Disease, Type C/blood , Niemann-Pick Disease, Type C/complications , Purkinje Cells/metabolism , Staining and Labeling , Survival Analysis , beta-Cyclodextrins/administration & dosage
18.
Proc Natl Acad Sci U S A ; 111(41): 14894-9, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25267637

ABSTRACT

Patients with mucopolysaccharidosis type I (MPS I), a genetic deficiency of the lysosomal enzyme α-l-iduronidase (IDUA), exhibit accumulation of glycosaminoglycans in tissues, with resulting diverse clinical manifestations including neurological, ocular, skeletal, and cardiac disease. MPS I is currently treated with hematopoietic stem cell transplantation or weekly enzyme infusions, but these therapies have significant drawbacks for patient safety and quality of life and do not effectively address some of the most critical clinical sequelae, such as life-threatening cardiac valve involvement. Using the naturally occurring feline model of MPS I, we tested liver-directed gene therapy as a means of achieving long-term systemic IDUA reconstitution. We treated four MPS I cats at 3-5 mo of age with an adeno-associated virus serotype 8 vector expressing feline IDUA from a liver-specific promoter. We observed sustained serum enzyme activity for 6 mo at ∼ 30% of normal levels in one animal, and in excess of normal levels in three animals. Remarkably, treated animals not only demonstrated reductions in glycosaminoglycan storage in most tissues, but most also exhibited complete resolution of aortic valve lesions, an effect that has not been previously observed in this animal model or in MPS I patients treated with current therapies. These data point to clinically meaningful benefits of the robust enzyme expression achieved with hepatic gene transfer that extend beyond the economic and quality of life advantages over lifelong enzyme infusions.


Subject(s)
Cardiovascular Diseases/therapy , Genetic Therapy , Liver/metabolism , Mucopolysaccharidosis I/therapy , Animals , Aortic Valve/metabolism , Aortic Valve/pathology , Cardiovascular Diseases/pathology , Cats , Dependovirus/genetics , Female , Genetic Vectors/metabolism , Glycosaminoglycans/metabolism , Heparin Cofactor II/metabolism , Iduronidase/blood , Iduronidase/genetics , Iduronidase/therapeutic use , Liver/pathology , Male , Molecular Sequence Data , Mucopolysaccharidosis I/blood , Mucopolysaccharidosis I/pathology , Myocardium/metabolism , Myocardium/pathology , Thrombin/metabolism , Tissue Distribution , Transduction, Genetic
19.
Mol Ther ; 22(12): 2018-2027, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25027660

ABSTRACT

Enzyme replacement therapy has revolutionized the treatment of the somatic manifestations of lysosomal storage diseases (LSD), although it has been ineffective in treating central nervous system (CNS) manifestations of these disorders. The development of neurotrophic vectors based on novel serotypes of adeno-associated viruses (AAV) such as AAV9 provides a potential platform for stable and efficient delivery of enzymes to the CNS. We evaluated the safety and efficacy of intrathecal delivery of AAV9 expressing α-l-iduronidase (IDUA) in a previously described feline model of mucopolysaccharidosis I (MPS I). A neurological phenotype has not been defined in these animals, so our analysis focused on the biochemical and histological CNS abnormalities characteristic of MPS I. Five MPS I cats were dosed with AAV9 vector at 4-7 months of age and followed for 6 months. Treated animals demonstrated virtually complete correction of biochemical and histological manifestations of the disease throughout the CNS. There was a range of antibody responses against IDUA in this cohort which reduced detectable enzyme without substantially reducing efficacy; there was no evidence of toxicity. This first demonstration of the efficacy of intrathecal gene therapy in a large animal model of a LSD should pave the way for translation into the clinic.


Subject(s)
Cats , Central Nervous System/pathology , Disease Models, Animal , Genetic Therapy/methods , Iduronidase/blood , Iduronidase/cerebrospinal fluid , Mucopolysaccharidosis I/therapy , Animals , Dependovirus/enzymology , Dependovirus/genetics , Genetic Vectors/administration & dosage , Injections, Spinal , Mucopolysaccharidosis I/enzymology , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/pathology , Organ Specificity
20.
J Bone Miner Res ; 29(12): 2610-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24898323

ABSTRACT

Mucopolysaccharidosis I (MPS I) is a lysosomal storage disease characterized by deficient α-L-iduronidase activity, leading to the accumulation of poorly degraded glycosaminoglycans (GAGs). Children with MPS I exhibit high incidence of spine disease, including accelerated disc degeneration and vertebral dysplasia, which in turn lead to spinal cord compression and kyphoscoliosis. In this study we investigated the efficacy of neonatal enzyme replacement therapy (ERT), alone or in combination with oral simvastatin (ERT + SIM) for attenuating cervical spine disease progression in MPS I, using a canine model. Four groups were studied: normal controls; MPS I untreated; MPS I ERT-treated; and MPS I ERT + SIM-treated. Animals were euthanized at age 1 year. Intervertebral disc condition and spinal cord compression were evaluated from magnetic resonance imaging (MRI) images and plain radiographs, vertebral bone condition and odontoid hypoplasia were evaluated using micro-computed tomography (µCT), and epiphyseal cartilage to bone conversion was evaluated histologically. Untreated MPS I animals exhibited more advanced disc degeneration and more severe spinal cord compression than normal animals. Both treatment groups resulted in partial preservation of disc condition and cord compression, with ERT + SIM not significantly better than ERT alone. Untreated MPS I animals had significantly lower vertebral trabecular bone volume and mineral density, whereas ERT treatment resulted in partial preservation of these properties. ERT + SIM treatment demonstrated similar, but not greater, efficacy. Both treatment groups partially normalized endochondral ossification in the vertebral epiphyses (as indicated by absence of persistent growth plate cartilage), and odontoid process size and morphology. These results indicate that ERT begun from a very early age attenuates the severity of cervical spine disease in MPS I, particularly for the vertebral bone and odontoid process, and that additional treatment with simvastatin does not provide a significant additional benefit over ERT alone.


Subject(s)
Dog Diseases , Enzyme Replacement Therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Iduronidase/therapeutic use , Intervertebral Disc Degeneration , Mucopolysaccharidosis I , Simvastatin/pharmacology , Animals , Disease Models, Animal , Dog Diseases/drug therapy , Dog Diseases/metabolism , Dog Diseases/pathology , Dogs , Female , Intervertebral Disc Degeneration/drug therapy , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Degeneration/veterinary , Male , Mucopolysaccharidosis I/drug therapy , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/pathology , Mucopolysaccharidosis I/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL
...