Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Ther ; 27(7): 1242-1251, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31056400

ABSTRACT

Citrin deficiency is an autosomal recessive disorder caused by loss-of-function mutations in SLC25A13, encoding the liver-specific mitochondrial aspartate/glutamate transporter. It has a broad spectrum of clinical phenotypes, including life-threatening neurological complications. Conventional protein replacement therapy is not an option for these patients because of drug delivery hurdles, and current gene therapy approaches (e.g., AAV) have been hampered by immunogenicity and genotoxicity. Although dietary approaches have shown some benefits in managing citrin deficiency, the only curative treatment option for these patients is liver transplantation, which is high-risk and associated with long-term complications because of chronic immunosuppression. To develop a new class of therapy for citrin deficiency, codon-optimized mRNA encoding human citrin (hCitrin) was encapsulated in lipid nanoparticles (LNPs). We demonstrate the efficacy of hCitrin-mRNA-LNP therapy in cultured human cells and in a murine model of citrin deficiency that resembles the human condition. Of note, intravenous (i.v.) administration of the hCitrin-mRNA resulted in a significant reduction in (1) hepatic citrulline and blood ammonia levels following oral sucrose challenge and (2) sucrose aversion, hallmarks of hCitrin deficiency. In conclusion, mRNA-LNP therapy could have a significant therapeutic effect on the treatment of citrin deficiency and other mitochondrial enzymopathies with limited treatment options.


Subject(s)
Citrullinemia/drug therapy , Citrullinemia/metabolism , Drug Delivery Systems/methods , Genetic Therapy/methods , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , RNA, Messenger/therapeutic use , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Gene Knockout Techniques , Glucosephosphate Dehydrogenase/genetics , HeLa Cells , Hep G2 Cells , Humans , Lipids/chemistry , Loss of Function Mutation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Nanoparticles/chemistry , Open Reading Frames/genetics , RNA, Messenger/chemical synthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Transfection , Treatment Outcome
2.
Am J Hum Genet ; 104(4): 625-637, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30879639

ABSTRACT

Fabry disease is an X-linked lysosomal storage disease caused by loss of alpha galactosidase A (α-Gal A) activity and is characterized by progressive accumulation of globotriaosylceramide and its analogs in all cells and tissues. Although enzyme replacement therapy (ERT) is considered standard of care, the long-term effects of ERT on renal and cardiac manifestations remain uncertain and thus novel therapies are desirable. We herein report preclinical studies evaluating systemic messenger RNA (mRNA) encoding human α-Gal A in wild-type (WT) mice, α-Gal A-deficient mice, and WT non-human primates (NHPs). The pharmacokinetics and distribution of h-α-Gal A mRNA encoded protein in WT mice demonstrated prolonged half-lives of α-Gal A in tissues and plasma. Single intravenous administration of h-α-Gal A mRNA to Gla-deficient mice showed dose-dependent protein activity and substrate reduction. Moreover, long duration (up to 6 weeks) of substrate reductions in tissues and plasma were observed after a single injection. Furthermore, repeat i.v. administration of h-α-Gal A mRNA showed a sustained pharmacodynamic response and efficacy in Fabry mice model. Lastly, multiple administrations to non-human primates confirmed safety and translatability. Taken together, these studies across species demonstrate preclinical proof-of-concept of systemic mRNA therapy for the treatment of Fabry disease and this approach may be useful for other lysosomal storage disorders.


Subject(s)
Fabry Disease/genetics , Fabry Disease/therapy , RNA, Messenger/therapeutic use , alpha-Galactosidase/genetics , Animals , Disease Models, Animal , Endocytosis , Enzyme Replacement Therapy , Genetic Therapy , Humans , Lipids/chemistry , Lysosomes/metabolism , Macaca fascicularis , Male , Mice , Mice, Knockout , RNA, Messenger/pharmacokinetics , Tissue Distribution , Trihexosylceramides/metabolism
3.
Nat Med ; 24(12): 1899-1909, 2018 12.
Article in English | MEDLINE | ID: mdl-30297912

ABSTRACT

Acute intermittent porphyria (AIP) results from haploinsufficiency of porphobilinogen deaminase (PBGD), the third enzyme in the heme biosynthesis pathway. Patients with AIP have neurovisceral attacks associated with increased hepatic heme demand. Phenobarbital-challenged mice with AIP recapitulate the biochemical and clinical characteristics of patients with AIP, including hepatic overproduction of the potentially neurotoxic porphyrin precursors. Here we show that intravenous administration of human PBGD (hPBGD) mRNA (encoded by the gene HMBS) encapsulated in lipid nanoparticles induces dose-dependent protein expression in mouse hepatocytes, rapidly normalizing urine porphyrin precursor excretion in ongoing attacks. Furthermore, hPBGD mRNA protected against mitochondrial dysfunction, hypertension, pain and motor impairment. Repeat dosing in AIP mice showed sustained efficacy and therapeutic improvement without evidence of hepatotoxicity. Finally, multiple administrations to nonhuman primates confirmed safety and translatability. These data provide proof-of-concept for systemic hPBGD mRNA as a potential therapy for AIP.


Subject(s)
Genetic Therapy , Hydroxymethylbilane Synthase/genetics , Porphyria, Acute Intermittent/therapy , RNA, Messenger/administration & dosage , Animals , Disease Models, Animal , Female , Haploinsufficiency/genetics , Heme/genetics , Heme/metabolism , Hepatocytes/drug effects , Humans , Hydroxymethylbilane Synthase/therapeutic use , Liver/drug effects , Liver/metabolism , Male , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/pathology , RNA, Messenger/genetics
5.
Mol Ther ; 26(6): 1509-1519, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29653760

ABSTRACT

The success of mRNA-based therapies depends on the availability of a safe and efficient delivery vehicle. Lipid nanoparticles have been identified as a viable option. However, there are concerns whether an acceptable tolerability profile for chronic dosing can be achieved. The efficiency and tolerability of lipid nanoparticles has been attributed to the amino lipid. Therefore, we developed a new series of amino lipids that address this concern. Clear structure-activity relationships were developed that resulted in a new amino lipid that affords efficient mRNA delivery in rodent and primate models with optimal pharmacokinetics. A 1-month toxicology evaluation in rat and non-human primate demonstrated no adverse events with the new lipid nanoparticle system. Mechanistic studies demonstrate that the improved efficiency can be attributed to increased endosomal escape. This effort has resulted in the first example of the ability to safely repeat dose mRNA-containing lipid nanoparticles in non-human primate at therapeutically relevant levels.


Subject(s)
Lipids/chemistry , Nanoparticles/chemistry , RNA, Messenger/administration & dosage , RNA, Messenger/chemistry , Animals , Primates , Rats
6.
Cell Rep ; 21(12): 3548-3558, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29262333

ABSTRACT

Isolated methylmalonic acidemia/aciduria (MMA) is a devastating metabolic disorder with poor outcomes despite current medical treatments. Like other mitochondrial enzymopathies, enzyme replacement therapy (ERT) is not available, and although promising, AAV gene therapy can be limited by pre-existing immunity and has been associated with genotoxicity in mice. To develop a new class of therapy for MMA, we generated a pseudoU-modified codon-optimized mRNA encoding human methylmalonyl-CoA mutase (hMUT), the enzyme most frequently mutated in MMA, and encapsulated it into biodegradable lipid nanoparticles (LNPs). Intravenous (i.v.) administration of hMUT mRNA in two different mouse models of MMA resulted in a 75%-85% reduction in plasma methylmalonic acid and was associated with increased hMUT protein expression and activity in liver. Repeat dosing of hMUT mRNA reduced circulating metabolites and dramatically improved survival and weight gain. Additionally, repeat i.v. dosing did not increase markers of liver toxicity or inflammation in heterozygote MMA mice.


Subject(s)
Amino Acid Metabolism, Inborn Errors/therapy , Genetic Therapy/methods , Methylmalonyl-CoA Mutase/genetics , Nanoparticles/administration & dosage , RNA, Messenger/genetics , Administration, Intravenous , Animals , Female , Humans , Lipids/chemistry , Liver/metabolism , Male , Methylmalonyl-CoA Mutase/metabolism , Mice , Nanoparticles/chemistry , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...