Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 13(1): 5012, 2022 08 25.
Article in English | MEDLINE | ID: mdl-36008405

ABSTRACT

Conventional therapy for hereditary tyrosinemia type-1 (HT1) with 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC) delays and in some cases fails to prevent disease progression to liver fibrosis, liver failure, and activation of tumorigenic pathways. Here we demonstrate cure of HT1 by direct, in vivo administration of a therapeutic lentiviral vector targeting the expression of a human fumarylacetoacetate hydrolase (FAH) transgene in the porcine model of HT1. This therapy is well tolerated and provides stable long-term expression of FAH in pigs with HT1. Genomic integration displays a benign profile, with subsequent fibrosis and tumorigenicity gene expression patterns similar to wild-type animals as compared to NTBC-treated or diseased untreated animals. Indeed, the phenotypic and genomic data following in vivo lentiviral vector administration demonstrate comparative superiority over other therapies including ex vivo cell therapy and therefore support clinical application of this approach.


Subject(s)
Precancerous Conditions , Tyrosinemias , Animals , Disease Models, Animal , Genetic Therapy , Humans , Hydrolases/genetics , Hydrolases/metabolism , Liver Cirrhosis/therapy , Nitrobenzoates/pharmacology , Nitrobenzoates/therapeutic use , Swine , Tyrosinemias/genetics , Tyrosinemias/therapy
2.
J Inherit Metab Dis ; 44(6): 1369-1381, 2021 11.
Article in English | MEDLINE | ID: mdl-33896013

ABSTRACT

Phenylketonuria (PKU) is the most common inborn error of metabolism of the liver, and results from mutations of both alleles of the phenylalanine hydroxylase gene (PAH). As such, it is a suitable target for gene therapy via gene delivery with a recombinant adeno-associated virus (AAV) vector. Here we use the synthetic AAV vector Anc80 via systemic administration to deliver a functional copy of a codon-optimized human PAH gene, with or without an intron spacer, to the Pahenu2 mouse model of PKU. Dose-dependent transduction of the liver and expression of PAH mRNA were present with both vectors, resulting in significant and durable reduction of circulating phenylalanine, reaching near control levels in males. Coat color of treated Pahenu2 mice reflected an increase in pigmentation from brown to the black color of control animals, further indicating functional restoration of phenylalanine metabolism and its byproduct melanin. There were no adverse effects associated with administration of AAV up to 5 × 1012 VG/kg, the highest dose tested. Only minor and/or transient variations in some liver enzymes were observed in some of the AAV-dosed animals which were not associated with pathology findings in the liver. Finally, there was no impact on cell turnover or apoptosis as evaluated by Ki-67 and TUNEL staining, further supporting the safety of this approach. This study demonstrates the therapeutic potential of AAV Anc80 to safely and durably cure PKU in a mouse model, supporting development for clinical consideration.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Phenylalanine Hydroxylase/genetics , Phenylketonurias/therapy , Animals , Cell Line , DNA, Recombinant/administration & dosage , Disease Models, Animal , Female , Genetic Vectors/genetics , Hair Color , Humans , Injections, Intravenous , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Phenylalanine/blood , Phenylalanine Hydroxylase/immunology , Phenylalanine Hydroxylase/metabolism , Transduction, Genetic/methods
3.
PLoS One ; 16(1): e0245831, 2021.
Article in English | MEDLINE | ID: mdl-33493163

ABSTRACT

Phenylketonuria (PKU) is a metabolic disorder whereby phenylalanine metabolism is deficient due to allelic variations in the gene for phenylalanine hydroxylase (PAH). There is no cure for PKU other than orthotopic liver transplantation, and the standard of care for patients is limited to dietary restrictions and key amino acid supplementation. Therefore, Pah was edited in pig fibroblasts for the generation of PKU clone piglets that harbor a common and severe human mutation, R408W. Additionally, the proximal region to the mutation was further humanized by introducing 5 single nucleotide polymorphisms (SNPs) to allow for development of gene editing machinery that could be translated directly from the pig model to human PKU patients that harbor at least one classic R408W allele. Resulting piglets were hypopigmented (a single Ossabaw piglet) and had low birthweight (all piglets). The piglets had similar levels of PAH expression, but no detectable enzymatic activity, consistent with the human phenotype. The piglets were fragile and required extensive neonatal care to prevent failure to thrive and early demise. Phenylalanine levels rose sharply when dietary Phe was unrestricted but could be rapidly reduced with a low Phe diet. Fibroblasts isolated from R408W piglets show susceptibility to correction using CRISPR or TALEN, with subsequent homology-directed recombination to correct Pah. This pig model of PKU provides a powerful new tool for development of all classes of therapeutic candidates to treat or cure PKU, as well as unique value for proof-of-concept studies for in vivo human gene editing platforms in the context of this humanized PKU allele.


Subject(s)
Gene Editing/methods , Mutation , Phenylalanine Hydroxylase/genetics , Phenylketonurias/genetics , Animals , Base Sequence , Disease Models, Animal , Humans , Phenotype , Safety , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...