Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Hum Gene Ther ; 33(11-12): 577-578, 2022 06.
Article in English | MEDLINE | ID: mdl-35537480
3.
Mol Ther ; 26(2): 496-509, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29292164

ABSTRACT

Hemophilia A is an X-linked bleeding disorder caused by mutations in the gene encoding the factor VIII (FVIII) coagulation protein. Bleeding episodes in patients are reduced by prophylactic therapy or treated acutely using recombinant or plasma-derived FVIII. We have made an adeno-associated virus 5 vector containing a B domain-deleted (BDD) FVIII gene (BMN 270) with a liver-specific promoter. BMN 270 injected into hemophilic mice resulted in a dose-dependent expression of BDD FVIII protein and a corresponding correction of bleeding time and blood loss. At the highest dose tested, complete correction was achieved. Similar corrections in bleeding were observed at approximately the same plasma levels of FVIII protein produced either endogenously by BMN 270 or following exogenous administration of recombinant BDD FVIII. No evidence of liver dysfunction or hepatocyte endoplasmic reticulum stress was observed. Comparable doses in primates produced similar levels of circulating FVIII. These preclinical data support evaluation of BMN 270 in hemophilia A patients.


Subject(s)
Factor VIII/genetics , Genetic Therapy , Hemophilia A/genetics , Hemophilia A/therapy , Peptide Fragments/genetics , Animals , Apoptosis/genetics , Cell Line , Dependovirus/genetics , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Gene Expression , Gene Order , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hemophilia A/blood , Liver/metabolism , Male , Mice , Mice, Transgenic , Peptide Fragments/blood , Primates , Promoter Regions, Genetic
5.
Hum Gene Ther ; 28(4): 323-327, 2017 04.
Article in English | MEDLINE | ID: mdl-27832705

ABSTRACT

There are conflicting reports that integration of the wild-type adeno-associated virus 2 (AAV2) genome is associated with induction of hepatocellular carcinoma (HCC) in a small subset of patients. However, there are several lines of evidence that contradict this assertion: (i) AAV2 has long been known to be a non-pathogenic virus, although ∼90% of the human population is seropositive for AAV2 antibodies; (ii) AAV2 has been shown to possess anticancer activity; (iii) epidemiological evidence suggests that AAV2 infection plays a protective role against cervical carcinoma; and (iv) five different AAV serotype vectors (AAV1, AAV2, AAV5, AAV8, and AAV9) have been or are currently being used in 162 Phase I/II clinical trials and one Phase III clinical trial in humans to date, and no cancer of any type has ever been observed or reported. A brief historical account of the putative role of infection by AAV in the etiology of cancer, or lack thereof, is presented.


Subject(s)
Carcinoma, Hepatocellular/therapy , Dependovirus/genetics , Genetic Therapy , Liver Neoplasms/therapy , Animals , Carcinoma, Hepatocellular/genetics , Clinical Trials as Topic , Dependovirus/classification , Gene Transfer Techniques , Genetic Vectors/therapeutic use , Humans , Liver Neoplasms/genetics , Mice
8.
Antioxid Redox Signal ; 11(7): 1569-86, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19187001

ABSTRACT

Enhanced redox-stress caused by neuroinflammation, mitochondria, and NADPH oxidases has been hypothesized to play critical roles in disease progression of amyotrophic lateral sclerosis (ALS). However, distinguishing whether the redox-stress observed in ALS is due to a primary defect in cellular reactive oxygen species metabolism/catabolism, or is a secondary consequence of neuroinflammation, has been difficult and the issue remains a matter of debate. Emerging evidence suggests that defects in genes that regulate NADPH oxidases may account for at least some forms of ALS. NADPH oxidases are key signaling complexes that influence cellular responses to growth factors and cytokines. In this context, NADPH oxidase-derived reactive oxygen species exert spatial control over the redox-dependent activation of certain pro-inflammatory receptors. Understanding the biology of how NADPH oxidases control cell signaling may help to clarify how genetic determinants of ALS lead to dysregulated pro-inflammatory signaling. This review provides a framework for understanding endosomal signaling through NADPH oxidases and potential mechanisms whereby gene defects in various forms of ALS may influence this cellular process and lead to motor neuron degeneration. Lastly, this review discusses past and current efforts to treat ALS using antioxidant therapies, as well as the limitations and advantages of each of these approaches.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/enzymology , Animals , Disease Models, Animal , Disease Progression , Humans , Mice , Mice, Transgenic , NADPH Oxidases/metabolism , Oxidation-Reduction , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
9.
Mol Ther ; 17(6): 1053-63, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19240687

ABSTRACT

Huntington's disease (HD) is a fatal neurodegenerative disease caused by mutant huntingtin (htt) protein, and there are currently no effective treatments. Recently, we and others demonstrated that silencing mutant htt via RNA interference (RNAi) provides therapeutic benefit in HD mice. We have since found that silencing wild-type htt in adult mouse striatum is tolerated for at least 4 months. However, given the role of htt in various cellular processes, it remains unknown whether nonallele-specific silencing of both wild-type and mutant htt is a viable therapeutic strategy for HD. Here, we tested whether cosilencing wild-type and mutant htt provides therapeutic benefit and is tolerable in HD mice. After treatment, HD mice showed significant reductions in wild-type and mutant htt, and demonstrated improved motor coordination and survival. We performed transcriptional profiling to evaluate the effects of reducing wild-type htt in adult mouse striatum. We identified gene expression changes that are concordant with previously described roles for htt in various cellular processes. Also, several abnormally expressed transcripts associated with early-stage HD were differentially expressed in our studies, but intriguingly, those involved in neuronal function changed in opposing directions. Together, these encouraging and surprising findings support further testing of nonallele-specific RNAi therapeutics for HD.


Subject(s)
Gene Silencing/physiology , Huntington Disease/therapy , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Line , Gene Expression Regulation/genetics , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/physiology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Plasmids , Polymerase Chain Reaction , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology
10.
Proc Natl Acad Sci U S A ; 105(15): 5868-73, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18398004

ABSTRACT

Huntington's disease (HD) is a fatal, dominant neurodegenerative disease caused by a polyglutamine repeat expansion in exon 1 of the HD gene, which encodes the huntingtin protein. We and others have shown that RNAi is a candidate therapy for HD because expression of inhibitory RNAs targeting mutant human HD transgenes improved neuropathology and behavioral deficits in HD mouse models. Here, we developed shRNAs targeting conserved sequences in human HD and mouse HD homolog (HDh) mRNAs to initiate preclinical testing in a knockin mouse model of HD. We screened 35 shRNAs in vitro and subsequently narrowed our focus to three candidates for in vivo testing. Unexpectedly, two active shRNAs induced significant neurotoxicity in mouse striatum, although HDh mRNA expression was reduced to similar levels by all three. Additionally, a control shRNA containing mismatches also induced toxicity, although it did not reduce HDh mRNA expression. Interestingly, the toxic shRNAs generated higher antisense RNA levels, compared with the nontoxic shRNA. These results demonstrate that the robust levels of antisense RNAs emerging from shRNA expression systems can be problematic in the mouse brain. Importantly, when sequences that were toxic in the context of shRNAs were placed into artificial microRNA (miRNA) expression systems, molecular and neuropathological readouts of neurotoxicity were significantly attenuated without compromising mouse HDh silencing efficacy. Thus, miRNA-based approaches may provide more appropriate biological tools for expressing inhibitory RNAs in the brain, the implications of which are crucial to the development of RNAi for both basic biological and therapeutic applications.


Subject(s)
MicroRNAs/pharmacology , Neurotoxicity Syndromes/drug therapy , RNA Interference , RNA, Small Interfering/adverse effects , Animals , Brain/drug effects , Corpus Striatum , Gene Silencing , Genetic Therapy/methods , Humans , Huntington Disease/therapy , Mice , MicroRNAs/chemical synthesis , MicroRNAs/therapeutic use , Neurotoxicity Syndromes/etiology
11.
N Engl J Med ; 358(21): 2231-9, 2008 May 22.
Article in English | MEDLINE | ID: mdl-18441371

ABSTRACT

Early-onset, severe retinal dystrophy caused by mutations in the gene encoding retinal pigment epithelium-specific 65-kD protein (RPE65) is associated with poor vision at birth and complete loss of vision in early adulthood. We administered to three young adult patients subretinal injections of recombinant adeno-associated virus vector 2/2 expressing RPE65 complementary DNA (cDNA) under the control of a human RPE65 promoter. There were no serious adverse events. There was no clinically significant change in visual acuity or in peripheral visual fields on Goldmann perimetry in any of the three patients. We detected no change in retinal responses on electroretinography. One patient had significant improvement in visual function on microperimetry and on dark-adapted perimetry. This patient also showed improvement in a subjective test of visual mobility. These findings provide support for further clinical studies of this experimental approach in other patients with mutant RPE65. (ClinicalTrials.gov number, NCT00643747 [ClinicalTrials.gov].).


Subject(s)
Blindness/therapy , Carrier Proteins/genetics , Eye Proteins/genetics , Genetic Therapy , Genetic Vectors , Retinal Degeneration/therapy , Adolescent , Adult , Blindness/congenital , Blindness/genetics , Blindness/pathology , DNA, Complementary , Dependovirus/genetics , Gene Transfer Techniques , Humans , Injections , Mutation , Retina/pathology , Retina/physiopathology , Retinal Degeneration/congenital , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Visual Acuity , cis-trans-Isomerases
12.
Hum Gene Ther ; 18(8): 726-32, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17685853

ABSTRACT

Previous studies have demonstrated that delivery of a recombinant adeno-associated virus (AAV) vector encoding the complete human cystic fibrosis transmembrane regulator (CFTR) cDNA (tgAAVCF) to the nose, sinus, and lungs of subjects with cystic fibrosis (CF) was safe and well tolerated. In a small randomized, double-blind study of three doses of aerosolized tgAAVCF or placebo at 30-day intervals, encouraging but non-significant trends in pulmonary function and induced sputum interleukin 8 (IL-8) levels were seen at early time points. This larger study was conducted to verify these trends. One hundred and two subjects aged 12 years and older with mild-to-moderate cystic fibrosis (forced expiratory flow in 1 sec [FEV1]:60% predicted) were randomized to two aerosolized doses of 1x10(13)DNase-resistant particles of tgAAVCF (n=51) or matching placebo (n=51) administered 30 days apart. Although tgAAVCF was well tolerated, the study did not meet its primary efficacy end point of statistically significant improvement in FEV1 30 days after initial administration of tgAAVCF compared with placebo. There were no significant differences in spirometric lung function over time, induced sputum biologic markers, or days of antibiotic use in either treatment group. Thus repeated doses of aerosolized tgAAVCF were safe and well tolerated, but did not result in significant improvement in lung function over time. Because gene transfer is the simplest, most basic way to correct the underlying genetic defect that leads to disease in CF, further research is warranted to develop an effective gene transfer agent for the treatment of CF.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/therapy , Dependovirus , Genetic Therapy , Administration, Inhalation , Adult , Child , Female , Humans , Male , Placebos
13.
Hum Gene Ther ; 16(8): 921-8, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16076250

ABSTRACT

Recombinant adeno-associated virus serotype 2 (rAAV2)-based human gene therapy for cystic fibrosis has progressed through a series of preclinical studies and phase I and II clinical trials. This agent has shown an encouraging safety profile, consistent levels of DNA transfer, and positive evidence of short-term clinical improvement in lung function in a prospective, placebo-controlled phase II trial of aerosol administration. Nonetheless, it has been difficult to assess the relationship between its molecular action and the observed clinical improvements, because of the lack of positive results from a highly specific assay for vector mRNA. This issue is further complicated by the fact that the clinical vector utilizes a small cryptic rAAV2 promoter sequence that is less robust for mRNA expression than typical viral promoters. In this paper, we report the results of more sensitive assays performed on primary nasal cells harvested from rAAV2-CFTR gene therapy recipients. These studies demonstrate a correlation between the presence of rAAV2-CFTR vector genomes, CFTR mRNA expression, and cAMP-activated chloride channel function in these cells. The observation of sizeable physiological correction in the face of low mRNA levels may reflect the regulatory role of low levels of CFTR protein as an activator of other chloride channels.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/physiopathology , Cystic Fibrosis/therapy , DNA, Viral/analysis , Epithelial Cells/physiology , Gene Transfer Techniques , Genetic Therapy , Adenoviridae , Administration, Intranasal , Blotting, Southern , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/analysis , Gene Expression Profiling , Genetic Vectors , Humans , Nasal Cavity/cytology , Polymerase Chain Reaction , RNA, Messenger , Sensitivity and Specificity
16.
Mol Ther ; 10(6): 990-1002, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15564131

ABSTRACT

Pharmacologic- and gene-based therapies have historically been developed as two independent therapeutic platforms for cystic fibrosis (CF) lung disease. Inhibition of the dysregulated epithelial Na channel (ENaC) is one pharmacologic approach to enhance airway clearance in CF. We investigated pharmacologic approaches to enhance CFTR gene delivery with recombinant adeno-associated virus (rAAV) and identified compounds that significantly improved viral transduction while simultaneously inhibiting ENaC activity through an unrelated mechanism. Treatment of human CF airway epithelia with proteasome modulating agents (LLnL and doxorubicin) at the time of rAAV2 or rAAV2/5 infection dramatically enhanced CFTR gene delivery and correction of CFTR-mediated short-circuit currents. Surprisingly, these agents also facilitated long-term (15-day) functional inhibition of ENaC currents independent of CFTR vector administration. Inhibition of ENaC activity was predominantly attributed to a doxorubicin-dependent decrease in gamma-ENaC subunit mRNA expression and an increase in gamma-ENaC promoter methylation. This is the first report to describe the identification of compounds with dual therapeutic action that are able to enhance the efficacy of CFTR gene therapy to the airway while simultaneously ameliorating primary aspects of CF disease pathophysiology. The identification of such compounds mark a new area for drug development, not only for CF, but also for other gene therapy disease targets.


Subject(s)
Cystic Fibrosis/genetics , Cystic Fibrosis/therapy , Doxorubicin/pharmacology , Genetic Therapy/methods , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Amiloride/pharmacology , Cell Polarity , Cells, Cultured , CpG Islands/genetics , Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , DNA Methylation , Dependovirus/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Sodium Channels , Genome, Viral , Humans , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium/metabolism , Sodium Channels/genetics , Sodium Channels/metabolism , Time Factors
17.
Mol Ther ; 9(2): 231-40, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14759807

ABSTRACT

The successful application of gene therapy for the treatment of genetic diseases such as Fabry is reliant on the development of vectors that are safe and that facilitate sustained expression of therapeutic levels of the transgene product. Here, we report that intravenous administration of a recombinant AAV2 vector encoding human alpha-galactosidase A under the transcriptional control of a liver-restricted enhancer/promoter (AAV2/DC190-alphagal) generated significantly higher levels of expression in BALB/c and Fabry mice than could be realized using the ubiquitous CMV promoter (AAV2/CMVHI-alphagal). Moreover, AAV2/DC190-alphagal-mediated hepatic expression of alpha-galactosidase A was sustained for 12 months in BALB/c mice and was associated with a significantly reduced immune response to the expressed enzyme. Subsequent challenge of the AAV2/DC190-alphagal-treated animals with recombinant human alpha-galactosidase A at 6 months failed to elicit the production of anti-alpha-galactosidase A antibodies, suggesting the induction of immune tolerance in these animals. The levels of expression attained with AAV2/DC190-alphagal in the Fabry mice were sufficient to reduce the abnormal accumulation of globotriaosylceramide in the liver, spleen, and heart to basal levels and in the kidney by approximately 40% at 8 weeks. Together, these results demonstrate that AAV2-mediated gene transfer that limits the expression of alpha-galactosidase A to the liver may be a viable strategy for treating Fabry disease.


Subject(s)
Dependovirus/genetics , Fabry Disease/therapy , Genetic Therapy , Immune Tolerance , Liver/metabolism , Promoter Regions, Genetic/genetics , alpha-Galactosidase/therapeutic use , Animals , DNA, Recombinant/genetics , Disease Models, Animal , Enhancer Elements, Genetic/genetics , Fabry Disease/genetics , Genetic Engineering , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism
19.
Hum Gene Ther ; 14(11): 1079-88, 2003 Jul 20.
Article in English | MEDLINE | ID: mdl-12885347

ABSTRACT

Recombinant adeno-associated serotype 2-based vectors (rAAV2) possess a number of theoretical advantages for cystic fibrosis (CF) gene therapy because they elicit little or no inflammatory response and generally result in stable expression. rAAV2 vectors expressing the cystic fibrosis transmembrane conductance regulator (CFTR) gene have previously been shown to mediate stable correction of the CF defect in CF bronchial epithelial cells and stable expression of CFTR in rabbit and nonhuman primate models. Here we report the results of the first trial initiated with rAAV in humans, a phase I study in 25 adult and adolescent CF patients with mild to moderate lung disease. Doses of the rAAV-CFTR vector (tgAAVCF) ranging from 3 x 10(1) to 1 x 10(9) replication units (RU), which is equivalent to approximately 6 x 10(4) to 2 x 10(12) DNase resistant particles (DRP), were administered to one side of the nose and to the superior segment of the lower lobe of the right lung. Several adverse events were noted prior to and/or after vector delivery, but most of them appeared to be related to the endogenous CF lung disease or a result of the bronchoscopic procedures. Only one of the serious events was judged to be possibly vector-related (based on temporal association), and this event was a pulmonary exacerbation very similar to several others experienced by the same subject in the three months preceding vector delivery. Vector shedding was minimal throughout the study, and serum-neutralizing antibodies were detected after vector delivery to subjects in the highest dosage cohorts. Gene transfer as measured by DNA polymerase chain reaction (PCR) was not observed until cohort 10 in nasal and bronchial epithelia. Sporadic low-level copy numbers suggested gene transfer of anywhere from 0.002 copies per cell up to 0.5 copies per cell was possible; however, DNA PCR was positive in lungs prior to direct dosing suggesting aspiration from the nasal dosing. These data indicate the need for continued evaluation of rAAV-CFTR vectors in additional clinical trials.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/therapy , Dependovirus/genetics , Genetic Vectors/toxicity , Administration, Intranasal , Adolescent , Adult , Antibodies, Viral/blood , Bronchi , Cystic Fibrosis/diagnosis , Cystic Fibrosis/immunology , DNA, Recombinant/genetics , DNA, Viral/analysis , Dependovirus/immunology , Enzyme-Linked Immunosorbent Assay , Genetic Vectors/administration & dosage , Humans , Interleukins/biosynthesis , Male , Viremia/diagnosis , Virus Shedding
SELECTION OF CITATIONS
SEARCH DETAIL
...