Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Inherit Metab Dis ; 41(6): 977-984, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29802554

RESUMO

BACKGROUND: Viral mediated gene therapy has progressed after overcoming early failures, and gene therapy has now been approved for several conditions in Europe and the USA. Glycogen storage disease (GSD) type Ia, caused by a deficiency of glucose-6-phosphatase-α, has been viewed as an outstanding candidate for gene therapy. This follow-up report describes the long-term outcome for the naturally occurring GSD-Ia dogs treated with rAAV-GPE-hG6PC-mediated gene therapy. METHODS: A total of seven dogs were treated with rAAV-GPE-hG6PC-mediated gene therapy. The first four dogs were treated at birth, and three dogs were treated between 2 and 6 months of age to assess the efficacy and safety in animals with mature livers. Blood and urine samples, radiographic studies, histological evaluation, and biodistribution were assessed. RESULTS: Gene therapy improved survival in the GSD-Ia dogs. With treatment, the biochemical studies normalized for the duration of the study (up to 7 years). None of the rAAV-GPE-hG6PC-treated dogs had focal hepatic lesions or renal abnormalities. Dogs treated at birth required a second dose of rAAV after 2-4 months; gene therapy after hepatic maturation resulted in improved efficacy after a single dose. CONCLUSION: rAAV-GPE-hG6PC treatment in GSD-Ia dogs was found to be safe and efficacious. GSD-Ia is an attractive target for human gene therapy since it is a monogenic disorder with limited tissue involvement. Blood glucose and lactate monitoring can be used to assess effectiveness and as a biomarker of success. GSD-Ia can also serve as a model for other hepatic monogenic disorders.


Assuntos
Terapia Genética/métodos , Doença de Depósito de Glicogênio Tipo I/terapia , Animais , Glicemia/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Cães , Europa (Continente) , Vetores Genéticos , Glucose-6-Fosfatase/genética , Hipoglicemia/genética , Hipoglicemia/metabolismo , Rim/metabolismo , Fígado/metabolismo
2.
Hum Mol Genet ; 20(R1): R61-8, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21518733

RESUMO

Pompe disease is an autosomal recessive metabolic myopathy caused by the deficiency of the lysosomal enzyme acid alpha-glucosidase and results in cellular lysosomal and cytoplasmic glycogen accumulation. A wide spectrum of disease exists from hypotonia and severe cardiac hypertrophy in the first few months of life due to severe mutations to a milder form with the onset of symptoms in adulthood. In either condition, the involvement of several systems leads to progressive weakness and disability. In early-onset severe cases, the natural history is characteristically cardiorespiratory failure and death in the first year of life. Since the advent of enzyme replacement therapy (ERT), the clinical outcomes have improved. However, it has become apparent that a new natural history is being defined in which some patients have substantial improvement following ERT, while others develop chronic disability reminiscent of the late-onset disease. In order to improve on the current clinical outcomes in Pompe patients with diminished clinical response to ERT, we sought to address the cause and potential for the treatment of disease manifestations which are not amenable to ERT. In this review, we will focus on the preclinical studies that are relevant to the development of a gene therapy strategy for Pompe disease, and have led to the first clinical trial of recombinant adeno-associated virus-mediated gene-based therapy for Pompe disease. We will cover the preliminary laboratory studies and rationale for a clinical trial, which is based on the treatment of the high rate of respiratory failure in the early-onset patients receiving ERT.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Doença de Depósito de Glicogênio Tipo II/terapia , Ensaios Clínicos como Assunto , Terapia de Reposição de Enzimas , Vetores Genéticos/administração & dosagem , Glicogênio/metabolismo , Doença de Depósito de Glicogênio Tipo II/imunologia , Doença de Depósito de Glicogênio Tipo II/patologia , Humanos , Resultado do Tratamento
3.
Sci Transl Med ; 15(677): eabo1815, 2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36599002

RESUMO

Duchenne muscular dystrophy (DMD) is a progressive muscle wasting disease caused by the absence of dystrophin, a membrane-stabilizing protein encoded by the DMD gene. Although mouse models of DMD provide insight into the potential of a corrective therapy, data from genetically homologous large animals, such as the dystrophin-deficient golden retriever muscular dystrophy (GRMD) model, may more readily translate to humans. To evaluate the clinical translatability of an adeno-associated virus serotype 9 vector (AAV9)-microdystrophin (µDys5) construct, we performed a blinded, placebo-controlled study in which 12 GRMD dogs were divided among four dose groups [control, 1 × 1013 vector genomes per kilogram (vg/kg), 1 × 1014 vg/kg, and 2 × 1014 vg/kg; n = 3 each], treated intravenously at 3 months of age with a canine codon-optimized microdystrophin construct, rAAV9-CK8e-c-µDys5, and followed for 90 days after dosing. All dogs received prednisone (1 milligram/kilogram) for a total of 5 weeks from day -7 through day 28. We observed dose-dependent increases in tissue vector genome copy numbers; µDys5 protein in multiple appendicular muscles, the diaphragm, and heart; limb and respiratory muscle functional improvement; and reduction of histopathologic lesions. As expected, given that a truncated dystrophin protein was generated, phenotypic test results and histopathologic lesions did not fully normalize. All administrations were well tolerated, and adverse events were not seen. These data suggest that systemically administered AAV-microdystrophin may be dosed safely and could provide therapeutic benefit for patients with DMD.


Assuntos
Distrofia Muscular Animal , Distrofia Muscular de Duchenne , Animais , Cães , Humanos , Recém-Nascido , Camundongos , Distrofina/genética , Distrofina/metabolismo , Terapia Genética , Coração , Músculo Esquelético/metabolismo , Músculos/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/terapia , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia
4.
Genet Vaccines Ther ; 10(1): 3, 2012 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-22709483

RESUMO

BACKGROUND: The appropriate tropism of adeno-associated virus (AAV) vectors that are systemically injected is crucial for successful gene therapy when local injection is not practical. Acidic oligopeptides have been shown to enhance drug delivery to bones. METHODS: In this study six-L aspartic acids (D6) were inserted into the AAV2 capsid protein sequence between amino acid residues 587 and 588. 129SVE mice were injected with double-stranded wild-type- (WT-) or D6-AAV2 mCherry expression vectors (3.24 x 1010 vg per animal) via the superficial temporal vein within 24 hours of birth. RESULTS: Fluorescence microscopy and quantitative polymerase chain reaction confirmed higher levels of mCherry expression in the paraspinal and gluteus muscles in the D6-AAV2 injected mice. The results revealed that although D6-AAV2 was less efficient in the transduction of immortalized cells stronger mCherry signals were detected over the spine and pelvis by live imaging in the D6-AAV2-injected mice than were detected in the WT-AAV2-injected mice. In addition, D6-AAV2 lost the liver tropism observed for WT-AAV2. CONCLUSIONS: An acidic oligopeptide displayed on AAV2 improves axial muscle tropism and decreases liver tropism after systemic delivery. This modification should be useful in creating AAV vectors that are suitable for gene therapy for diseases involving the proximal muscles.

5.
Proc Natl Acad Sci U S A ; 106(38): 16363-8, 2009 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-19706466

RESUMO

Alpha-1 antitrypsin (AAT) deficiency is well-suited as a target for human gene transfer. We performed a phase 1, open-label, dose-escalation clinical trial of a recombinant adeno-associated virus (rAAV) vector expressing normal (M) AAT packaged into serotype 1 AAV capsids delivered by i.m. injection. Nine AAT-deficient subjects were enrolled sequentially in cohorts of 3 each at doses of 6.9 x 10(12), 2.2 x 10(13), and 6.0 x 10(13) vector genome particles per patient. Four subjects receiving AAT protein augmentation discontinued therapy 28 or 56 days before vector administration. Vector administration was well tolerated, with only mild local reactions and 1 unrelated serious adverse event (bacterial epididymitis). There were no changes in hematology or clinical chemistry parameters. M-specific AAT was expressed above background in all subjects in cohorts 2 and 3 and was sustained at levels 0.1% of normal for at least 1 year in the highest dosage level cohort, despite development of neutralizing antibody and IFN-gamma enzyme-linked immunospot responses to AAV1 capsid at day 14 in all subjects. These findings suggest that immune responses to AAV capsid that develop after i.m. injection of a serotype 1 rAAV vector expressing AAT do not completely eliminate transduced cells in this context.


Assuntos
Terapia Genética/métodos , Linfócitos T/metabolismo , Deficiência de alfa 1-Antitripsina/terapia , alfa 1-Antitripsina/metabolismo , Adulto , Idoso , Anticorpos Antivirais/sangue , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Capsídeo/enzimologia , Capsídeo/imunologia , Linhagem Celular , Dependovirus/genética , Dependovirus/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Expressão Gênica , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Injeções Intramusculares , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes de Fusão/sangue , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Fatores de Tempo , alfa 1-Antitripsina/sangue , alfa 1-Antitripsina/genética , Deficiência de alfa 1-Antitripsina/genética
6.
J Biomed Biotechnol ; 2011: 646257, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21318173

RESUMO

A canine model of Glycogen storage disease type Ia (GSDIa) is described. Affected dogs are homozygous for a previously described M121I mutation resulting in a deficiency of glucose-6-phosphatase-α. Metabolic, clinicopathologic, pathologic, and clinical manifestations of GSDIa observed in this model are described and compared to those observed in humans. The canine model shows more complete recapitulation of the clinical manifestations seen in humans including "lactic acidosis", larger size, and longer lifespan compared to other animal models. Use of this model in preclinical trials of gene therapy is described and briefly compared to the murine model. Although the canine model offers a number of advantages for evaluating potential therapies for GSDIa, there are also some significant challenges involved in its use. Despite these challenges, the canine model of GSDIa should continue to provide valuable information about the potential for generating curative therapies for GSDIa as well as other genetic hepatic diseases.


Assuntos
Modelos Animais de Doenças , Doenças do Cão/genética , Doenças do Cão/metabolismo , Doença de Depósito de Glicogênio Tipo I/genética , Doença de Depósito de Glicogênio Tipo I/metabolismo , Hepatopatias/genética , Hepatopatias/metabolismo , Animais , Ensaios Clínicos como Assunto , Cães , Doença de Depósito de Glicogênio Tipo I/diagnóstico , Doença de Depósito de Glicogênio Tipo I/patologia , Doença de Depósito de Glicogênio Tipo I/veterinária , Humanos , Hepatopatias/veterinária
7.
Mol Ther ; 18(3): 502-10, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20104213

RESUMO

Pompe disease is a muscular dystrophy that results in respiratory insufficiency. We characterized the outcomes of targeted delivery of recombinant adeno-associated virus serotype 1 (rAAV2/1) vector to diaphragms of Pompe mice with varying stages of disease progression. We observed significant improvement in diaphragm contractile strength in mice treated at 3 months of age that is sustained at least for 1 year and enhanced contractile strength in mice treated at 9 and 21 months of age, measured 3 months post-treatment. Ventilatory parameters including tidal volume/inspiratory time ratio, minute ventilation/expired CO2 ratio, and peak inspiratory airflow were significantly improved in mice treated at 3 months and tested at 6 months. Despite early improvement, mice treated at 3 months and tested at 1 year had diminished normoxic ventilation, potentially due to attenuation of correction over time or progressive degeneration of nontargeted accessory tissues. However, for all rAAV2/1-treated mice (treated at 3, 9, and 21 months, assayed 3 months later; treated at 3 months, assayed at 1 year), minute ventilation and peak inspiratory flows were significantly improved during respiratory challenge. These results demonstrate that gel-mediated delivery of rAAV2/1 vectors can significantly augment ventilatory function at initial and late phases of disease in a model of muscular dystrophy.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Doença de Depósito de Glicogênio Tipo II/genética , Doença de Depósito de Glicogênio Tipo II/terapia , Distrofias Musculares/terapia , Respiração , Animais , Dióxido de Carbono/química , Progressão da Doença , Géis , Vetores Genéticos , Camundongos , Camundongos Transgênicos , Contração Muscular , Distrofias Musculares/genética , Fatores de Tempo
8.
Sci Rep ; 10(1): 3930, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32127618

RESUMO

A splice site mutation in the canine pyruvate dehydrogenase kinase 4 (PDK4) gene has been shown to be associated with the development of dilated cardiomyopathy (DCM) in Doberman Pinchers (DPs). Subsequent studies have successfully demonstrated the use of dermal fibroblasts isolated from DPs as models for PDK4 deficiency and have shown activation of the intrinsic (mitochondrial mediated) apoptosis pathway in these cells under starvation conditions. For this study, we sought to further explore the functional consequences of PDK4 deficiency in DP fibroblasts representing PDK4wt/wt, PDK4wt/del, and PDK4del/del genotypes. Our results show that starvation conditions cause increased perinuclear localization of mitochondria and decreased cell proliferation, altered expression levels of pyruvate dehydrogenase phosphatase (PDP) and pyruvate dehydrogenase (PDH), dramatically increased PDH activity, and an impaired response to mitochondrial stress in affected cells. In sum, these results show the broad impact of PDK4 deficiency and reveal mechanistic pathways used by these cells in an attempt to compensate for the condition. Our data help to elucidate the mechanisms at play in this extremely prevalent DP disorder and provide further support demonstrating the general importance of metabolic flexibility in cell health.


Assuntos
Fibroblastos/enzimologia , Proteínas Quinases/deficiência , Western Blotting , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Microscopia de Fluorescência , Consumo de Oxigênio/genética , Consumo de Oxigênio/fisiologia , Fosforilação/genética , Fosforilação/fisiologia , Proteínas Quinases/genética , Piruvato Desidrogenase (Lipoamida)-Fosfatase/genética , Piruvato Desidrogenase (Lipoamida)-Fosfatase/metabolismo , Complexo Piruvato Desidrogenase/genética , Complexo Piruvato Desidrogenase/metabolismo
9.
Circ Res ; 99(4): e3-9, 2006 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-16873720

RESUMO

Heart disease is often the end result of inherited genetic defects, which may potentially be treatable using a gene-transfer approach. Recombinant adeno-associated virus (rAAV)-mediated gene delivery has emerged as a realistic method for the treatment of such disorders. Here, we demonstrate and compare the natural affinity of specific AAV serotype capsids for transduction of cardiac tissue. We compared the previously accepted optimal rAAV serotype for transduction of skeletal muscle, rAAV2/1, with rAAV2/8 and the newer rAAV2/9 vectors carrying the CMV-lacZ construct in their respective abilities to transcend vasculature and transduce myocardium following intravenous delivery of 1x10(11) vector genomes in neonatal mice. We found that both rAAV2/8 and rAAV2/9 are able to transduce myocardium at approximately 20- and 200-fold (respectively) higher levels than rAAV2/1. Biodistribution analysis revealed that rAAV2/9 and rAAV2/8 demonstrate similar behavior in extracardiac tissue. Vector genome quantification showed an increase in genome copy numbers in cardiac tissue for several weeks following administration, which corresponds to expression data. In addition, we intravenously administered 1x10(11) vector genomes of rAAV2/9-CMV-lacZ into adult mice and achieved an expression biodistribution profile similar to that found following delivery to newborns. Although higher doses of virus will be necessary to approach those levels observed following neonatal injections, adult myocardium is also readily transduced by rAAV2/9. Finally, we have demonstrated physiological disease correction by AAV9 gene transfer in a mouse model of Pompe disease via ECG tracings and that intravenous delivery of the same vector preferentially transduces cardiac tissue in nonhuman primates.


Assuntos
Dependovirus/genética , Dependovirus/patogenicidade , Coração/virologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Eletrocardiografia , Genes Reporter , Vetores Genéticos , Haplorrinos , Camundongos , Recombinação Genética , Sorotipagem , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
10.
J Huntingtons Dis ; 7(4): 309-319, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30320596

RESUMO

BACKGROUND: Transgenic sheep are currently the only large animal model of Huntington's disease expressing full-length mutant human huntingtin. These transgenic sheep provide an opportunity to test adeno associated virus (AAV) therapies directly targeting the huntingtin gene. A recent study demonstrated that self-complementary (sc) AAV with artificial miRNA against human huntingtin reduced mutant human huntingtin in caudate and putamen after a single injection near the internal capsule. OBJECTIVE: To identify an AAV serotype among AAVrh8, AAV9 and AAVrh10 with the highest neuronal uptake and distribution, with no obvious cell loss in the neostriatum of the sheep. METHODS: We tested AAVrh8, AAV9 and AAVrh10 by stereotactic direct unilateral injection into the neostriatum of sheep, near the internal capsule. Four weeks after administration, we examined the viral spread and neuronal uptake of each serotype of AAV containing GFP. We compared single stranded (ss) and scAAVs. Further, we measured the distribution of AAVrh8 and AAV9 to a variety of tissues outside the brain. RESULTS: Sc AAV9 had the best combination of neuronal uptake and distribution throughout the neostriatum. scAAVrh10 demonstrated good spread, but was not taken up by neurons. scAAVrh8 demonstrated good spread, but had less neuronal uptake than AAV9. Six hours after convection-enhanced administration to the neostriatum, both AAVrh8 and AAV9 viral genomes were detected in blood, saliva, urine, feces and wool. By four weeks, viral genomes were detected in wool only. Administration of AAVrh8, AAV9 and AAVrh10 was not associated with loss of neostriatal, medium spiny neuron number as measured by DARPP32 immunohistochemistry. CONCLUSIONS: Altogether, we found scAAV9 had the best neuronal uptake and spread, showed no loss of neurons at one-month post-injection, and was not measurable in body fluids one month after injection. This information will guide future clinical experiments requiring brain injection of AAV for therapeutics for gene or miRNA deliveries in sheep transgenic for the human huntingtin gene.


Assuntos
Núcleo Caudado/virologia , Dependovirus/genética , Proteína Huntingtina/genética , Neurônios/virologia , Putamen/virologia , Internalização do Vírus , Animais , Animais Geneticamente Modificados , Dependovirus/metabolismo , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos/sangue , Vetores Genéticos/urina , Genoma Viral , Proteínas de Fluorescência Verde/genética , Humanos , Cápsula Interna , Masculino , Neostriado/virologia , Sorogrupo , Ovinos , Carneiro Doméstico , Lã/virologia
11.
Hum Gene Ther Clin Dev ; 29(4): 188-197, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30280954

RESUMO

Applied Genetic Technologies Corporation (AGTC) is developing a recombinant adeno-associated virus (rAAV) vector AGTC-501, also designated AAV2tYF-GRK1-RPGRco, to treat retinitis pigmentosa (RP) in patients with mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene. The vector contains a codon-optimized human RPGR cDNA (RPGRco) driven by a photoreceptor-specific promoter (G protein-coupled receptor kinase 1, GRK1) and is packaged in an AAV2 capsid with three surface tyrosine residues changed to phenylalanine (AAV2tYF). We conducted a safety and potency study of this vector administered by subretinal a injection in the naturally occurring RPGR-deficient Rd9 mouse model. Sixty Rd9 mice (20 per group) received a subretinal injection in the right eye of vehicle (control) or AAV2tYF-GRK1-RPGRco at one of two dose levels (4 × 108 or 4 × 109 vg/eye) and were followed for 12 weeks after injection. Vector injections were well tolerated, with no systemic toxicity. There was a trend towards reduced electroretinography b-wave amplitudes in the high vector dose group that was not statistically significant. There were no clinically important changes in hematology or clinical chemistry parameters and no vector-related ocular changes in life or by histological examination. Dose-dependent RPGR protein expression, mainly in the inner segment of photoreceptors and the adjacent connecting cilium region, was observed in all vector-treated eyes examined. Sequence integrity of the codon-optimized RPGR was confirmed by sequencing of PCR-amplified DNA, or cDNA reverse transcribed from total RNA extracted from vector-treated retinal tissues, and by sequencing of RPGR protein obtained from transfected HEK 293 cells. These results support the use of rAAV2tYF-GRK1-RPGRco in clinical studies in patients with XLRP caused by RPGR mutations.


Assuntos
Proteínas de Transporte/genética , Dependovirus/genética , Proteínas do Olho/genética , Receptor Quinase 1 Acoplada a Proteína G/genética , Terapia Genética/métodos , Retinose Pigmentar/terapia , Animais , Proteínas de Transporte/metabolismo , Códon/genética , Códon/metabolismo , Dependovirus/metabolismo , Proteínas do Olho/metabolismo , Receptor Quinase 1 Acoplada a Proteína G/metabolismo , Terapia Genética/efeitos adversos , Camundongos , Retinose Pigmentar/genética
12.
Hum Gene Ther ; 18(3): 245-56, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17376008

RESUMO

To translate the potential advantages of recombinant adeno-associated virus type 1 (rAAV1) vectors into a clinical application for muscle-directed gene therapy for alpha1 -antitrypsin (AAT) deficiency, we performed safety studies in 170 C57BL/6 mice and 26 New Zealand White rabbits. A mouse toxicology study included 8 cohorts of 10 mice each (5 per sex). Mice were killed either 21 or 90 days after intramuscular injection of doses ranging up to 1x10(13)vector genomes (VG), equivalent to 4 x 10(14)VG/kg. A mouse biodistribution study was performed in 5 cohorts of 10 mice, receiving intramuscular injections at the same doses; as well as in a lower dose cohort (3 x 10(8) VG; equivalent to 1.2 x 10(10)VG/kg); and in 4 other cohorts (excluding the vehicle control) injected with identical doses intravenously. Finally, biodistribution was examined in rabbits, with serial collection of blood and semen, as well as terminal tissue collection. Two significant findings were present, both of which were dose dependent. First, inflammatory cell infiltrates were detected at the site of injection 21, 60, or 90 days after intramuscular injection of 1 x 10(13)VG. This was not associated with loss of transgene expression. Second, vector DNA sequences were detected in most animals, levels being highest with the highest doses and earliest time points. Vector DNA was also present in liver, spleen, kidneys, and a number of other organs, including the gonads of animals receiving the highest dose. Likewise, vector DNA was present in the semen of male rabbits at higher doses. The copy number of vector DNA in the blood and semen declined over time throughout the study. These two dose-dependent findings have served to guide to the design of a phase 1 human trial of rAAV1-AAT.


Assuntos
Dependovirus/genética , Vetores Genéticos/toxicidade , alfa 1-Antitripsina/genética , Animais , DNA Viral/análise , DNA Viral/farmacocinética , DNA Viral/toxicidade , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/farmacocinética , Genoma Viral , Humanos , Inflamação/induzido quimicamente , Inflamação/patologia , Injeções Intramusculares , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/química , Músculo Esquelético/patologia , Coelhos , Distribuição Tecidual , alfa 1-Antitripsina/análise , Deficiência de alfa 1-Antitripsina/terapia
14.
Genet Vaccines Ther ; 5: 13, 2007 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-18070352

RESUMO

Adeno-associated virus (AAV) has shown great promise as a gene transfer vector. However, the incubation time needed to attain significant levels of gene expression is often too long for some clinical applications. Self-complementary AAV (scAAV) enters the cell as double stranded DNA, eliminating the step of second-strand synthesis, proven to be the rate-limiting step for gene expression of single-stranded AAV (ssAAV). The aim of this study was to compare the efficiency of these two types of AAV vectors in the murine myocardium. Four day old CD-1 mice were injected with either of the two AAV constructs, both expressing GFP and packaged into the AAV1 capsid. The animals were held for 4, 6, 11 or 21 days, after which they were euthanized and their hearts were excised. Serial sections of the myocardial tissue were used for real-time PCR quantification of AAV genome copies and for confocal microscopy. Although we observed similar numbers of AAV genomes at each of the different time points present in both the scAAV and the ssAAV infected hearts, microscopic analysis showed expression of GFP as early as 4 days in animals injected with the scAAV, while little or no expression was observed with the ssAAV constructs until day 11. AAV transduction of murine myocardium is therefore significantly enhanced using scAAV constructs.

15.
Hum Gene Ther Clin Dev ; 28(3): 145-156, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28726496

RESUMO

Cystic fibrosis (CF) is an autosomal recessive disease that is potentially treatable by gene therapy. Since the identification of the gene encoding CF transmembrane conductance regulator, a number of preclinical and clinical trials have been conducted using the first generation of adeno-associated virus, AAV2. All these studies showed that AAV gene therapy for CF is safe, but clinical benefit was not clearly demonstrated. Thus, a new generation of AAV vectors based on other serotypes is needed to move the field forward. This study tested two AAV serotypes (AAV1 and AAV5) using a dual-luciferase reporter system with firefly and Renilla luciferase genes packaged into AAV1 or AAV5, respectively. Two male and two female Rhesus macaques were each instilled in their lungs with both serotypes using a Penn-Century microsprayer. Both AAV1 and AAV5 vector genomes were detected in all the lung samples when measured at the time of necropsy, 45 days after instillation. However, the vector genome number for AAV1 was at least 10-fold higher than for AAV5. Likewise, luciferase activity was also detected in the same samples at 45 days. AAV1-derived activity was not statistically greater than that derived from AAV5. These data suggest that gene transfer is greater for AAV1 than for AAV5 in macaque lungs. Serum neutralizing antibodies were increased dramatically against both serotypes but were less abundant with AAV1 than with AAV5. No adverse events were noted, again indicating that AAV gene therapy is safe. These results suggest that with more lung-tropic serotypes such as AAV1, new clinical studies of gene therapy using AAV are warranted.


Assuntos
Fibrose Cística/terapia , Dependovirus/genética , Terapia Genética/métodos , Luciferases/genética , Animais , Feminino , Técnicas de Transferência de Genes/efeitos adversos , Genes Reporter , Terapia Genética/efeitos adversos , Vetores Genéticos/genética , Luciferases/metabolismo , Macaca mulatta , Masculino
16.
Hum Gene Ther Clin Dev ; 28(4): 208-218, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29160099

RESUMO

A first-in-human trial of diaphragmatic gene therapy (AAV1-CMV-GAA) to treat respiratory and neural dysfunction in early-onset Pompe disease was conducted. The primary objective of this study was to assess the safety of rAAV1-CMV-hGAA vector delivered to the diaphragm muscle of Pompe disease subjects with ventilatory insufficiency. Safety was assessed by measurement of change in serum chemistries and hematology, urinalysis, and immune response to GAA and AAV, as well as change in level of health. The data demonstrate that the AAV treatment was safe and there were no adverse events related to the study agent. Adverse events related to the study procedure were observed in subjects with lower baseline neuromuscular function. All adverse events were resolved before the end of the study, except for one severe adverse event determined not to be related to either the study agent or the study procedure. In addition, an anti-capsid and anti-transgene antibody response was observed in all subjects who received rAAV1-CMV-hGAA, except for subjects who received concomitant immunomodulation to manage reaction to enzyme replacement therapy, as per their standard of care. This observation is significant for future gene therapy studies and serves to establish a clinically relevant approach to blocking immune responses to both the AAV capsid protein and transgene product.


Assuntos
Dependovirus/genética , Terapia Genética , Doença de Depósito de Glicogênio Tipo II/genética , alfa-Glucosidases/administração & dosagem , Animais , Criança , Diafragma/cirurgia , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/efeitos adversos , Doença de Depósito de Glicogênio Tipo II/sangue , Doença de Depósito de Glicogênio Tipo II/patologia , Doença de Depósito de Glicogênio Tipo II/terapia , Humanos , Imunomodulação , Masculino , Camundongos , Músculo Esquelético , Cirurgia Torácica Vídeoassistida , Transgenes/genética , alfa-Glucosidases/efeitos adversos , alfa-Glucosidases/genética
17.
Hum Gene Ther ; 17(1): 71-80, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16409126

RESUMO

Mitochondrial beta-oxidation of fatty acids is required to meet physiologic energy requirements during illness and periods of fasting or physiologic stress, and is most active in liver and striated muscle. Acyl-CoA dehydrogenases of varying chain-length specificities represent the first step in the mitochondria for each round of beta-oxidation, each of which removes two-carbon units as acetyl-CoA for entry into the tricarboxylic acid cycle. We have used recombinant adeno-associated virus (rAAV) vectors expressing short-chain acyl-CoA dehydrogenase (SCAD) to correct the accumulation of fatty acyl-CoA intermediates in deficient cell lines. The rAAV-SCAD vector was then packaged into either rAAV serotype 1 or 2 capsids and injected intramuscularly into SCAD-deficient mice. A systemic effect was observed as judged by restoration of circulating butyryl- carnitine levels to normal. Total lipid content at the injection site was also decreased as demonstrated by noninvasive magnetic resonance spectroscopy (MRS). SCAD enzyme activity in the injected muscle was found at necropsy to be above the normal control mouse level. This study is the first to demonstrate the systemic correction of a fatty acid oxidation disorder with rAAV and the utility of MRS as a noninvasive method to monitor SCAD correction after in vivo gene therapy.


Assuntos
Dependovirus/fisiologia , Ácidos Graxos/metabolismo , Terapia Genética/métodos , Vetores Genéticos/fisiologia , Erros Inatos do Metabolismo Lipídico/terapia , Animais , Butiril-CoA Desidrogenase/deficiência , Butiril-CoA Desidrogenase/genética , Butiril-CoA Desidrogenase/metabolismo , Carnitina/análogos & derivados , Carnitina/análise , Carnitina/sangue , Linhagem Celular , DNA Recombinante , Dependovirus/enzimologia , Ácidos Graxos/análise , Feminino , Fibroblastos/metabolismo , Humanos , Injeções Intramusculares , Espectrometria de Massas/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Músculos/química , Músculos/enzimologia , Oxirredução , Reprodutibilidade dos Testes , Transdução Genética
18.
Hum Gene Ther ; 17(12): 1177-86, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17115945

RESUMO

A phase I trial of intramuscular injection of a recombinant adeno-associated virus serotype 2 (rAAV2) alpha1-antitrypsin (AAT) vector was performed in 12 AAT-deficient adults, 10 of whom were male. All subjects were either homozygous for the most common AAT mutation (a missense mutation designated PI*Z) or compound heterozygous for PI*Z and another mutation known to cause disease. There were four dose cohorts, ranging from 2.1 x 10(12) vector genomes (VG) to 6.9 x 10(13) VG, with three subjects per cohort. Subjects were injected sequentially in a dose-escalating fashion with a minimum of 14 days between patients. Subjects who had been receiving AAT protein replacement discontinued that therapy 28 days before vector administration. There were no vector-related serious adverse events in any of the 12 participants. Vector DNA sequences were detected in the blood between 1 and 3 days after injection in nearly all patients receiving doses of 6.9 x 10(12) VG or higher. Anti-AAV2 capsid antibodies were present and rose after vector injection, but no other immune responses were detected. One subject who had not been receiving protein replacement exhibited low-level expression of wild-type M-AAT in the serum (82 nM), which was detectable 30 days after receiving an injection of 2.1 x 10(13) VG. Unfortunately, residual but declining M-AAT levels from the washout of the protein replacement elevated background levels sufficiently to obscure any possible vector expression in that range in most of the other individuals in the higher dose cohorts.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Deficiência de alfa 1-Antitripsina/terapia , alfa 1-Antitripsina/genética , Adulto , Idoso , DNA Recombinante/sangue , Dependovirus/classificação , Dependovirus/imunologia , Feminino , Terapia Genética/efeitos adversos , Vetores Genéticos , Humanos , Injeções Intramusculares , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , alfa 1-Antitripsina/sangue , Deficiência de alfa 1-Antitripsina/genética , Deficiência de alfa 1-Antitripsina/imunologia
19.
Hum Gene Ther ; 17(8): 845-58, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16942444

RESUMO

Leber congenital amaurosis (LCA) is a molecularly heterogeneous disease group that leads to blindness. LCA caused by RPE65 mutations has been studied in animal models and vision has been restored by subretinal delivery of AAV-RPE65 vector. Human ocular gene transfer trials are being considered. Our safety studies of subretinal AAV-2/2.RPE65 in RPE65-mutant dogs showed evidence of modest photoreceptor loss in the injection region in some animals at higher vector doses. We now test the hypothesis that there can be vectorrelated toxicity to the normal monkey, with its human-like retina. Good Laboratory Practice safety studies following single intraocular injections of AAV-2/2.RPE65 in normal cynomolgus monkeys were performed for 1-week and 3-month durations. Systemic toxicity was not identified. Ocular-specific studies included clinical examinations, electroretinography, and retinal histopathology. Signs of ocular inflammation postinjection had almost disappeared by 1 week. At 3 months, electroretinography in vector-injected eyes was no different than in vehicle-injected control eyes or compared with presurgical recordings. Healed sites of retinal perforation from subretinal injections were noted clinically and by histopathology. Foveal architecture in subretinally injected eyes, vector or vehicle, could be abnormal. Morphometry of central retina showed no photoreceptor layer thickness abnormalities occurring in a dose-dependent manner. Vector sequences were present in the injected retina, vitreous, and optic nerve at 1 week but not consistently in the brain. At 3 months, there were no vector sequences in optic nerve and brain. The results allow for consideration of an upper range for no observed adverse effect level in future human trials of subretinal AAV-2/2.RPE65. The potential value of foveal treatment for LCA and other retinal degenerations warrants further research into how to achieve gene transfer without retinal injury from surgical detachment of the retina.


Assuntos
Cegueira/terapia , Dependovirus , Proteínas do Olho , Terapia Genética , Atrofia Óptica Hereditária de Leber/terapia , Animais , Cegueira/etiologia , Cegueira/genética , Cegueira/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/virologia , Proteínas de Transporte , Cães , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Humanos , Macaca fascicularis , Mutação , Atrofia Óptica Hereditária de Leber/complicações , Atrofia Óptica Hereditária de Leber/genética , Atrofia Óptica Hereditária de Leber/patologia , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Nervo Óptico/virologia , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patologia , Células Fotorreceptoras/virologia , Retina/metabolismo , Retina/patologia , Retina/virologia , cis-trans-Isomerases
20.
Am J Vet Res ; 77(2): 156-61, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27027709

RESUMO

OBJECTIVE: To compare mitochondrial oxygen consumption rate (OCR) of fibroblasts from Doberman Pinschers with and without dilated cardiomyopathy (DCM) and mutation of the gene for pyruvate dehydrogenase kinase isozyme 4 (PDK4) and to evaluate in vitro whether treatment with adeno-associated virus (AAV) vector (i.e., gene therapy) would alter metabolic efficiency. ANIMALS: 10 Doberman Pinschers screened for DCM and PDK4 mutation. PROCEDURES Fibroblasts were harvested from skin biopsy specimens obtained from Doberman Pinschers, and dogs were classified as without DCM or PDK4 mutation (n = 3) or with occult DCM and heterozygous (4) or homozygous (3) for PDK4 mutation. Fibroblasts were or were not treated with tyrosine mutant AAV type 2 vector containing PDK4 at multiplicities of infection of 1,000. Mitochondrial OCR was measured to evaluate mitochondrial metabolism. The OCR was compared among dog groups and between untreated and treated fibroblasts within groups. RESULTS: Mean ± SD basal OCR of fibroblasts from heterozygous (74 ± 8 pmol of O2/min) and homozygous (58 ± 12 pmol of O2/min) dogs was significantly lower than that for dogs without PDK4 mutation (115 ± 9 pmol of O2/min). After AAV transduction, OCR did not increase significantly in any group (mutation-free group, 121 ± 26 pmol of O2/min; heterozygous group, 88 ± 6 pmol of O2/min; homozygous group, 59 ± 3 pmol of O2/min). CONCLUSIONS AND CLINICAL RELEVANCE: Mitochondrial function was altered in skin fibroblasts of Doberman Pinschers with DCM and PDK4 mutation. Change in mitochondrial function after in vitro gene therapy at the multiplicities of infection used in this study was not significant.


Assuntos
Cardiomiopatia Dilatada/veterinária , Doenças do Cão/metabolismo , Fibroblastos/metabolismo , Mitocôndrias/metabolismo , Proteínas Quinases/metabolismo , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/terapia , Dependovirus , Doenças do Cão/genética , Doenças do Cão/terapia , Cães , Fibroblastos/patologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Terapia Genética , Vetores Genéticos , Consumo de Oxigênio/genética , Proteínas Quinases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA