Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Transl Med ; 14(627): eabc0700, 2022 01 12.
Article in English | MEDLINE | ID: mdl-35020410

ABSTRACT

Correction of enzymatic deficits in hepatocytes by systemic administration of a recombinant protein is a desired therapeutic goal for hepatic enzymopenic disorders such as acute intermittent porphyria (AIP), an inherited porphobilinogen deaminase (PBGD) deficiency. Apolipoprotein A-I (ApoAI) is internalized into hepatocytes during the centripetal transport of cholesterol. Here, we generated a recombinant protein formed by linking ApoAI to the amino terminus of human PBGD (rhApoAI-PBGD) in an attempt to transfer PBGD into liver cells. In vivo experiments showed that, after intravenous injection, rhApoAI-PBGD circulates in blood incorporated into high-density lipoprotein (HDL), penetrates into hepatocytes, and crosses the blood-brain barrier, increasing PBGD activity in both the liver and brain. Consistently, the intravenous administration of rhApoAI-PBGD or the hyperfunctional rApoAI-PBGD-I129M/N340S (rApoAI-PBGDms) variant efficiently prevented and abrogated phenobarbital-induced acute attacks in a mouse model of AIP. One month after a single intravenous dose of rApoAI-PBGDms, the protein was still detectable in the liver, and hepatic PBGD activity remained increased above control values. A long-lasting therapeutic effect of rApoAI-PBGDms was observed after either intravenous or subcutaneous administration. These data describe a method to deliver PBGD to hepatocytes with resulting enhanced hepatic enzymatic activity and protection against AIP attacks in rodent models, suggesting that the approach might be an effective therapy for AIP.


Subject(s)
Hydroxymethylbilane Synthase , Porphyria, Acute Intermittent , Animals , Disease Models, Animal , Genetic Therapy/methods , Hydroxymethylbilane Synthase/metabolism , Hydroxymethylbilane Synthase/therapeutic use , Mice , Porphyria, Acute Intermittent/drug therapy , Porphyria, Acute Intermittent/metabolism
2.
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
3.
Hum Mol Genet ; 27(21): 3688-3696, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30085095

ABSTRACT

A first-in-human gene therapy trial using a recombinant adeno-associated viral (rAAV) vector for acute intermittent porphyria (AIP) reveals that higher doses would be required to reach therapeutic levels of the porphobilinogen deaminase (PBGD) transgene. We developed a hyperfunctional PBGD protein to improve the therapeutic index without increasing vector dose. A consensus protein sequence from 12 mammal species was compared to the human PBGD sequence, and eight amino acids were selected. I291M and N340S variants showed the highest increase in enzymatic activity when expressed in prokaryotic and eukaryotic systems. In silico analysis indicates that isoleucine 291 to methionine and asparagine 340 to serine variants did not affect the active site of the enzyme. In vitro analysis indicated a synergistic interaction between these two substitutions that improve kinetic stability. Finally, full protection against a phenobarbital-induced attack was achieved in AIP mice after the administration of 1 × 1011 gc/kg of rAAV2/8-PBGD-I291M/N340S vector; three times lower than the dose required to achieve full protection with the control rAAV2/8-hPBGD vector. In conclusion, we have developed and characterized a hyperfunctional PBGD protein. The inclusion of this variant sequence in a rAAV2/8 vector allows the effective dose to be lowered in AIP mice.


Subject(s)
Genetic Therapy , Hydroxymethylbilane Synthase/metabolism , Hydroxymethylbilane Synthase/therapeutic use , Porphyria, Acute Intermittent/therapy , Animals , Computer Simulation , Disease Models, Animal , Hydroxymethylbilane Synthase/pharmacology , Kinetics , Male , Mammals/metabolism , Mice , Phenobarbital/toxicity , Protein Conformation , Sequence Analysis, Protein , Therapeutic Index
4.
Hum Gene Ther ; 24(12): 1007-17, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24070415

ABSTRACT

Acute intermittent porphyria (AIP) results from haplo-insufficient activity of porphobilinogen deaminase (PBGD) and is characterized clinically by life-threatening, acute neurovisceral attacks. To date, liver transplantation is the only curative option for AIP. The aim of the present preclinical nonhuman primate study was to determine the safety and transduction efficacy of an adeno-associated viral vector encoding PBGD (recombinant AAV serotype 5-codon-optimized human porphobilinogen deaminase, rAAV5-cohPBGD) administered intravenously as part of a safety program to start a clinical study in patients with AIP. Macaques injected with either 1 × 10(13) or 5 × 10(13) vector genomes/kg of clinical-grade rAAV5-cohPBGD were monitored by standardized clinical parameters, and vector shedding was analyzed. Liver transduction efficacy, biodistribution, vector integration, and histopathology at day 30 postvector administration were determined. There was no evidence of acute toxicity, and no adverse effects were observed. The vector achieved efficient and homogenous hepatocellular transduction, reaching transgenic PBGD expression levels equivalent to 50% of the naturally expressed PBGD mRNA. No cellular immune response was detected against the human PBGD or AAV capsid proteins. Integration site analysis in transduced liver cells revealed an almost random integration pattern supporting the good safety profile of rAAV5-cohPBGD. Together, data obtained in nonhuman primates indicate that rAAV5-cohPBGD represents a safe therapy to correct the metabolic defect present in AIP patients.


Subject(s)
Genetic Therapy , Haploinsufficiency/genetics , Hydroxymethylbilane Synthase/genetics , Porphyria, Acute Intermittent/therapy , Animals , Dependovirus , Genetic Vectors , Hepatocytes/metabolism , Humans , Hydroxymethylbilane Synthase/therapeutic use , Macaca , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/pathology , Tissue Distribution/genetics , Transduction, Genetic
5.
Best Pract Res Clin Gastroenterol ; 24(5): 593-605, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20955962

ABSTRACT

The porphyrias are predominantly inherited metabolic disorders, which result from a specific deficiency of one of the eight enzymes along the pathway of haem biosynthesis. Historically, they have been classified into hepatic and erythropoietic forms, based on the primary site of expression of the prevailing dysfunctional enzyme. From a clinical point of view, however, it is more convenient to subdivide them into acute and non-acute porphyrias, thereby primarily considering the potential occurrence of life-threatening acute neurovisceral attacks. Unrecognised or untreated, such an acute porphyric attack is associated with a significant mortality of up to 10%. The acute hepatic porphyrias comprise acute intermittent porphyria, variegate porphyria, hereditary coproporphyria, and δ-aminolevulinic acid dehydratase deficiency porphyria. Making a precise diagnosis may be difficult because the different types of porphyrias may show overlapping clinical and biochemical characteristics. To date, the therapeutic possibilities are limited and mainly symptomatic. In this overview we report on what is currently known about pathogenesis, clinic, diagnostics, and therapy of the acute hepatic porphyrias. We further point out actual and future challenges in the management of these diseases.


Subject(s)
Porphyrias, Hepatic/therapy , Animals , Carcinoma, Hepatocellular/etiology , Coproporphyria, Hereditary/genetics , Enzyme Replacement Therapy , Female , Genetic Therapy , Humans , Hydroxymethylbilane Synthase/therapeutic use , Liver Neoplasms/etiology , Liver Transplantation , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/metabolism , Porphyrias, Hepatic/classification , Porphyrias, Hepatic/complications , Porphyrias, Hepatic/diagnosis , Porphyrias, Hepatic/genetics , Pregnancy , Pregnancy Complications/therapy , Recombinant Proteins/therapeutic use , Renal Insufficiency/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...