Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Ther ; 26(1): 256-268, 2018 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-29033008

RESUMO

The aim of this study was the evaluation of the safety and efficacy of unilateral subretinal injection of the adeno-associated vector (AAV) serotypes 2 and 4 (AAV2/4) RPE65-RPE65 vector in patients with Leber congenital amaurosis (LCA) associated with RPE65 gene deficiency. We evaluated ocular and general tolerance and visual function up to 1 year after vector administration in the most severely affected eye in nine patients with retinal degeneration associated with mutations in the RPE65 gene. Patients received either low (1.22 × 1010 to 2 × 1010 vector genomes [vg]) or high (between 3.27 × 1010 and 4.8 × 1010 vg) vector doses. An ancillary study, in which six of the original nine patients participated, extended the follow-up period to 2-3.5 years. All patients showed good ophthalmological and general tolerance to the rAAV2/4-RPE65-RPE65 vector. We observed a trend toward improved visual acuity in patients with nystagmus, stabilization and improvement of the visual field, and cortical activation along visual pathways during fMRI analysis. OCT analysis after vector administration revealed no retinal thinning, except in cases of macular detachment. Our findings show that the rAAV2/4.RPE65.RPE65 vector was well tolerated in nine patients with RPE65-associated LCA. Efficacy parameters varied between patients during follow-up.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/genética , Amaurose Congênita de Leber/genética , cis-trans-Isomerases/genética , Adolescente , Adulto , Análise de Variância , Criança , Seguimentos , Terapia Genética/métodos , Humanos , Amaurose Congênita de Leber/diagnóstico , Amaurose Congênita de Leber/metabolismo , Amaurose Congênita de Leber/terapia , Imageamento por Ressonância Magnética , Tomografia de Coerência Óptica , Campos Visuais , Adulto Jovem , cis-trans-Isomerases/metabolismo
2.
Mol Ther ; 24(5): 867-76, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26857842

RESUMO

We previously reported that subretinal injection of AAV2/5 RK.cpde6ß allowed long-term preservation of photoreceptor function and vision in the rod-cone dysplasia type 1 (rcd1) dog, a large animal model of naturally occurring PDE6ß deficiency. The present study builds on these earlier findings to provide a detailed assessment of the long-term effects of gene therapy on the spatiotemporal pattern of retinal degeneration in rcd1 dogs treated at 20 days of age. We analyzed the density distribution of the retinal layers and of particular photoreceptor cells in 3.5-year-old treated and untreated rcd1 dogs. Whereas no rods were observed outside the bleb or in untreated eyes, gene transfer halted rod degeneration in all vector-exposed regions. Moreover, while gene therapy resulted in the preservation of cones, glial cells and both the inner nuclear and ganglion cell layers, no cells remained in vector-unexposed retinas, except in the visual streak. Finally, the retinal structure of treated 3.5-year-old rcd1 dogs was identical to that of unaffected 4-month-old rcd1 dogs, indicating near complete preservation. Our findings indicate that gene therapy arrests the degenerative process even if intervention is initiated after the onset of photoreceptor degeneration, and point to significant potential of this therapeutic approach in future clinical trials.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Terapia Genética/métodos , Degeneração Retiniana/terapia , Células Fotorreceptoras Retinianas Bastonetes/patologia , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/deficiência , Dependovirus/genética , Modelos Animais de Doenças , Cães , Vetores Genéticos/administração & dosagem , Humanos , Retina/fisiopatologia , Degeneração Retiniana/genética , Degeneração Retiniana/patologia
3.
Mol Ther ; 22(2): 265-277, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24091916

RESUMO

For the development of new therapies, proof-of-concept studies in large animal models that share clinical features with their human counterparts represent a pivotal step. For inherited retinal dystrophies primarily involving photoreceptor cells, the efficacy of gene therapy has been demonstrated in canine models of stationary cone dystrophies and progressive rod-cone dystrophies but not in large models of progressive cone-rod dystrophies, another important cause of blindness. To address the last issue, we evaluated gene therapy in the retinitis pigmentosa GTPase regulator interacting protein 1 (RPGRIP1)-deficient dog, a model exhibiting a severe cone-rod dystrophy similar to that seen in humans. Subretinal injection of AAV5 (n = 5) or AAV8 (n = 2) encoding the canine Rpgrip1 improved photoreceptor survival in transduced areas of treated retinas. Cone function was significantly and stably rescued in all treated eyes (18-72% of those recorded in normal eyes) up to 24 months postinjection. Rod function was also preserved (22-29% of baseline function) in four of the five treated dogs up to 24 months postinjection. No detectable rod function remained in untreated contralateral eyes. More importantly, treatment preserved bright- and dim-light vision. Efficacy of gene therapy in this large animal model of cone-rod dystrophy provides great promise for human treatment.


Assuntos
Proteínas do Olho/genética , Terapia Genética , Retinose Pigmentar/genética , Retinose Pigmentar/terapia , Animais , Animais Geneticamente Modificados , Dependovirus/genética , Modelos Animais de Doenças , Progressão da Doença , Cães , Expressão Gênica , Técnicas de Inativação de Genes , Ordem dos Genes , Técnicas de Transferência de Genes , Genes Reporter , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Humanos , Regiões Promotoras Genéticas , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Retinose Pigmentar/patologia , Transdução Genética , Resultado do Tratamento
4.
Mol Ther ; 20(11): 2019-30, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22828504

RESUMO

Defects in the ß subunit of rod cGMP phosphodiesterase 6 (PDE6ß) are associated with autosomal recessive retinitis pigmentosa (RP), a childhood blinding disease with early retinal degeneration and vision loss. To date, there is no treatment for this pathology. The aim of this preclinical study was to test recombinant adeno-associated virus (AAV)-mediated gene addition therapy in the rod-cone dysplasia type 1 (rcd1) dog, a large animal model of naturally occurring PDE6ß deficiency that strongly resembles the human pathology. A total of eight rcd1 dogs were injected subretinally with AAV2/5RK.cpde6ß (n = 4) or AAV2/8RK.cpde6ß (n = 4). In vivo and post-mortem morphological analysis showed a significant preservation of the retinal structure in transduced areas of both AAV2/5RK.cpde6ß- and AAV2/8RK.cpde6ß-treated retinas. Moreover, substantial rod-derived electroretinography (ERG) signals were recorded as soon as 1 month postinjection (35% of normal eyes) and remained stable for at least 18 months (the duration of the study) in treated eyes. Rod-responses were undetectable in untreated contralateral eyes. Most importantly, dim-light vision was restored in all treated rcd1 dogs. These results demonstrate for the first time that gene therapy effectively restores long-term retinal function and vision in a large animal model of autosomal recessive rod-cone dystrophy, and provide great promise for human treatment.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/deficiência , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Retinose Pigmentar/terapia , Animais , Dependovirus/genética , Modelos Animais de Doenças , Cães , Eletrorretinografia , Terapia Genética , Vetores Genéticos , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Recuperação de Função Fisiológica , Retina/metabolismo , Retina/patologia , Retina/fisiopatologia , Vasos Retinianos/patologia , Retinose Pigmentar/fisiopatologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transgenes , Resultado do Tratamento , Visão Ocular
5.
Exp Eye Res ; 93(4): 491-502, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21723863

RESUMO

Systemic delivery of recombinant adeno-associated virus (rAAV) vectors has recently been shown to cross the blood brain barrier in rodents and large animals and to efficiently target cells of the central nervous system. Such approach could be particularly interesting to treat lysosomal storage diseases or neurodegenerative disorders characterized by multiple organs injuries especially neuronal and retinal dysfunctions. However, the ability of rAAV vector to cross the blood retina barrier and to transduce retinal cells after systemic injection has not been precisely determined. In this study, gene transfer was investigated in the retina of neonatal and adult rats after intravenous injection of self-complementary (sc) rAAV serotype 1, 5, 6, 8, and 9 carrying a CMV-driven green fluorescent protein (GFP), by fluorescence fundus photography and histological examination. Neonatal rats injected with scAAV2/9 vector displayed the strongest GFP expression in the retina, within the retinal pigment epithelium (RPE) cells. Retinal tropism of scAAV2/9 vector was further assessed after systemic delivery in large animal models, i.e., dogs and cats. Interestingly, efficient gene transfer was observed in the RPE cells of these two large animal models following neonatal intravenous injection of the vector. The ability of scAAV2/9 to transduce simultaneously neurons in the central nervous system, and RPE cells in the retina, after neonatal systemic delivery, makes this approach potentially interesting for the treatment of infantile neurodegenerative diseases characterized by both neuronal and retinal damages.


Assuntos
Dependovirus/genética , Expressão Gênica/fisiologia , Técnicas de Transferência de Genes , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Retina/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Gatos , DNA Complementar , Cães , Feminino , Angiofluoresceinografia , Proteínas de Fluorescência Verde/imunologia , Injeções Intravenosas , Gravidez , Ratos , Ratos Sprague-Dawley , Transgenes
6.
Mol Ther ; 18(6): 1085-93, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20354505

RESUMO

In previous studies, we demonstrated that recombinant adeno-associated virus (rAAV)-mediated gene transfer of the doxycycline (Dox)-regulatable system allows for the regulation of erythropoietin (EPO) expression in the retina of nonhuman primates after intravenous or oral administration of Dox. In addition, it was shown that administrating different amounts of Dox resulted in a dose-response dynamic of transgene expression. Adeno-associated viral gene therapy has raised hope for the treatment of patients with Leber congenital amaurosis, caused by mutations in the retinal pigment epithelium (RPE)-specific gene RPE65. The preliminary results of three clinical trials suggest some improvement in visual function. However, further improvements might be necessary to optimize vision recovery and this means developing vectors able to generate transgene expression at physiological levels. The purpose of this study was to investigate the ability of the Dox-regulatable system to regulate retinal function in RPE65(-/-) Briard dogs. rAAV vectors expressing RPE65 under the control of either the TetOff and TetOn Dox-regulated promoters or the cytomegalovirus (CMV) constitutive promoter were generated and administered subretinally to seven RPE65-deficient dogs. We demonstrate that the induction and deinduction of retinal function, as assessed by electroretinography (ERG), can be achieved using a Dox-regulatable system, but do not lead to any recovery of vision.


Assuntos
Dependovirus/genética , Doxiciclina/farmacologia , Proteínas do Olho/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Vetores Genéticos , Retina/fisiopatologia , Transtornos da Visão/terapia , Animais , Cães , Transtornos da Visão/fisiopatologia
7.
Mol Ther ; 18(1): 151-60, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19888197

RESUMO

In the absence of an immune response from the host, intramuscular (IM) injection of recombinant adeno-associated virus (rAAV) results in the permanent expression of the transgene from mouse to primate models. However, recent gene transfer studies into animal models and humans indicate that the risk of transgene and/or capsid-specific immune responses occurs and depends on multiple factors. Among these factors, the route of delivery is important, although poorly addressed in large animal models. Here, we compare the IM and the drug-free regional intravenous (RI) deliveries of rAAV in nonhuman primate (NHP) skeletal muscle monitoring the host immune response toward the transgene. We show that IM is consistently associated with immunotoxicity and the destruction of the genetically modified myofibers, whereas RI allows the stable expression of the transgene. This has important implications for the design of clinical trials for gene transfer in skeletal muscle.


Assuntos
Dependovirus/genética , Dependovirus/imunologia , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Injeções Intravenosas/efeitos adversos , Músculo Esquelético/metabolismo , Transdução Genética/métodos , Animais , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos/genética , Imuno-Histoquímica , Hibridização In Situ , Injeções Intramusculares/efeitos adversos , Macaca , Masculino , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Mol Ther ; 17(3): 516-23, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19107120

RESUMO

Gene transfer to the retina using recombinant adeno-associated viral (rAAV) vectors has proven to be an effective option for the treatment of retinal degenerative diseases in several animal models and has recently advanced into clinical trials in humans. To date, intracellular trafficking of AAV vectors and subsequent capsid degradation has been studied only in vitro, but the fate of AAV particles in transduced cells following subretinal injection has yet to be elucidated. Using electron microscopy and western blot, we analyzed retinas of one primate and four dogs that had been subretinally injected with AAV2/4, -2/5, or -2/2 serotypes and that displayed efficient gene transfer over several years. We show that intact AAV particles are still present in retinal cells, for up to 6 years after successful gene transfer in these large animals. The persistence of intact vector particles in the target organ, several years postadministration, is totally unexpected and, therefore, represents a new and unanticipated safety issue to consider at a time when gene therapy clinical trials raise new immunological concerns.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Primatas/genética , Primatas/metabolismo , Retina/metabolismo , Vírion/genética , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Dependovirus/ultraestrutura , Cães , Vetores Genéticos/administração & dosagem , Microscopia Imunoeletrônica , Fatores de Tempo
9.
J Virol ; 82(16): 7875-85, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18524821

RESUMO

Recombinant adeno-associated virus (rAAV) vectors are capable of mediating long-term gene expression following administration to skeletal muscle. In rodent muscle, the vector genomes persist in the nucleus in concatemeric episomal forms. Here, we demonstrate with nonhuman primates that rAAV vectors integrate inefficiently into the chromosomes of myocytes and reside predominantly as episomal monomeric and concatemeric circles. The episomal rAAV genomes assimilate into chromatin with a typical nucleosomal pattern. The persistence of the vector genomes and gene expression for years in quiescent tissues suggests that a bona fide chromatin structure is important for episomal maintenance and transgene expression. These findings were obtained from primate muscles transduced with rAAV1 and rAAV8 vectors for up to 22 months after intramuscular delivery of 5 x 10(12) viral genomes/kg. Because of this unique context, our data, which provide important insight into in situ vector biology, are highly relevant from a clinical standpoint.


Assuntos
Cromatina/química , Dependovirus/genética , Regulação Viral da Expressão Gênica , Genoma Viral , Músculo Esquelético/metabolismo , Animais , Cromatina/metabolismo , Cromossomos , Epigênese Genética , Técnicas de Transferência de Genes , Vetores Genéticos , Histonas/metabolismo , Macaca , Modelos Biológicos , Músculo Esquelético/virologia , Nucleossomos/metabolismo , Transgenes
10.
Mol Vis ; 15: 349-61, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19223988

RESUMO

PURPOSE: To evaluate the RPGRIP1-deficient miniature longhaired dachshund (MLHD) dog as a potential candidate for gene therapy. METHODS: Six RPGRIP1-deficient MLHD dogs from our dog colony have been observed for two years using a variety of noninvasive procedures. These included bilateral full-field electroretinograms (ERG) to evaluate retinal function, fundus photographs to evaluate retinal vascularization, and optical coherence tomographs (OCT) to evaluate retinal thickness. We also performed histological examination of hematoxylin- and eosin-stained retinal sections as well as sections labeled in situ by the terminal dUTP nick end labeling (TUNEL) method. RESULTS: ERG findings showed that as early as 2 months of age, cone function was lost while rod function was preserved. However, by 9 months of age, both cone and rod functions could not be detected. Functional visual assessment based on the ability to avoid obstacles showed that vision was retained up to the age of 11 months. Both OCT and histopathology studies revealed a progressive thinning of the outer nuclear layer (ONL) over the first 2 years of age. TUNEL labeling identified apoptotic photoreceptor cell death as the cause of this thinning of the ONL. CONCLUSIONS: A treatment strategy should consist in initiating gene therapy as early as possible after birth to prevent or delay the loss of rod function. In the MLHD, successful subretinal delivery of a therapeutic vector is feasible at 2 months of age and may prevent or delay the loss of rod function.


Assuntos
Cegueira , Modelos Animais de Doenças , Cães/genética , Terapia Genética , Proteínas/genética , Retina/patologia , Animais , Animais Geneticamente Modificados , Apoptose , Cegueira/genética , Cegueira/patologia , Cegueira/terapia , Eletrorretinografia , Imunofluorescência , Fundo de Olho , Marcação In Situ das Extremidades Cortadas , Distribuição Normal , Proteínas/metabolismo , Retina/citologia , Retina/metabolismo , Células Fotorreceptoras Retinianas Cones/citologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Cones/patologia , Células Fotorreceptoras Retinianas Bastonetes/citologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/patologia , Vasos Retinianos , Tomografia de Coerência Óptica , Visão Ocular/genética
11.
Mol Ther ; 16(5): 916-23, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18388922

RESUMO

Recombinant adeno-associated virus (rAAV) vectors are among the most efficient gene delivery vehicles for gene transfer to the retina. This study evaluates the behavior of the rAAV8 serotype vector with regard to intraocular delivery in rats and dogs. Subretinal delivery of an AAV2/8.gfp vector results in efficient gene transfer in the retinal pigment epithelium (RPE), the photoreceptors and, surprisingly, in the cells of the inner nuclear layer as well as in ganglion cells. Most importantly, in dogs, gene transfer also occurred distal to the injection site in neurons of the lateral geniculate nucleus of the brain. Because green fluorescent protein (GFP) was detected along the visual pathway within the brain, we analyzed total DNA extracted from various brain slices using PCR. Vector sequences were detected in many parts of the brain, but chiefly in the contralateral hemisphere.


Assuntos
Encéfalo/metabolismo , Dependovirus/metabolismo , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Neurônios/metabolismo , Retina/metabolismo , Animais , Núcleo Celular/metabolismo , Cães , Proteínas de Fluorescência Verde/metabolismo , Epitélio Pigmentado Ocular/metabolismo , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar
12.
Mol Ther ; 16(7): 1291-1299, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28178483

RESUMO

We developed a drug-free regional intravenous (RI) delivery protocol of recombinant adeno-associated virus (rAAV) 1 and 8 to an entire limb in the nonhuman primate (NHP), and compared the results with those produced by intramuscular (IM) delivery of the same dose of vector. We show that RI delivery of both serotypes was remarkably well tolerated with no adverse side-effects. After IM, muscle transduction was restricted to the site of injection with a high number of vector copies per cell for rAAV1. In contrast, although RI delivery resulted in a lower vector copy per cell, it was detectable in the vast majority of muscles of the injected limb. The amounts of circulating infectious rAAV were similar for both serotypes and modes of delivery. At autopsy at up to 34 months after vector administration, similar biodistribution patterns were found for both vectors and for both modes of delivery, with numerous organs found to be positive for vector sequence when assayed using PCR and Southern blot. Altogether, we demonstrated that RI is a simple and efficient transduction protocol in NHPs, resulting in higher expression of the transgene with a lower number of vector genomes per cell. However, regardless of the mode of delivery, concerns continue to be raised by the presence of vector sequences detected at distant sites.

13.
Mol Ther ; 16(7): 1291-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18461055

RESUMO

We developed a drug-free regional intravenous (r.i.) delivery protocol of recombinant adeno-associated virus (rAAV) 1 and 8 to an entire limb in the nonhuman primate (NHP), and compared the results with those produced by intramuscular (i.m.) delivery of the same dose of vector. We show that r.i. delivery of both serotypes was remarkably well tolerated with no adverse side-effects. After i.m., muscle transduction was restricted to the site of injection with a high number of vector copies per cell for rAAV1. In contrast, although r.i. delivery resulted in a lower vector copy per cell, it was detectable in the vast majority of muscles of the injected limb. The amounts of circulating infectious rAAV were similar for both serotypes and modes of delivery. At autopsy at up to 34 months after vector administration, similar biodistribution patterns were found for both vectors and for both modes of delivery, with numerous organs found to be positive for vector sequence when assayed using PCR and Southern blot. Altogether, we demonstrated that r.i. is a simple and efficient transduction protocol in NHPs, resulting in higher expression of the transgene with a lower number of vector genomes per cell. However, regardless of the mode of delivery, concerns continue to be raised by the presence of vector sequences detected at distant sites.


Assuntos
Dependovirus , Vetores Genéticos/administração & dosagem , Músculo Esquelético , Transdução Genética/métodos , Animais , DNA Viral/sangue , Expressão Gênica , Vetores Genéticos/efeitos adversos , Vetores Genéticos/farmacocinética , Injeções Intramusculares/efeitos adversos , Injeções Intravenosas/efeitos adversos , Macaca fascicularis , Masculino , Transgenes
14.
Transplantation ; 85(9): 1351-6, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18475195

RESUMO

Regulatory T cells (Treg) have been identified as playing a pivotal role in the control of tolerance and in the suppression of pathologic immune responses in autoimmune diseases, transplantation, and graft-versus-host disease. Treg expanded ex vivo by dendritic cells could be potential reagents to promote antigen-specific tolerance in vivo. However, in vivo studies have been carried out mostly in rodents and will need validation in primates before clinical application. We characterized macaque dendritic cell derived either from bone marrow with and without prior CD34+ cell selection (BMDC), or from CD14+ peripheral blood mononuclear cells (Mo-DC). We demonstrate that with a semi-mature phenotype, BMDC are superior to Mo-DC in their capacity to expand freshly isolated allogeneic macaque CD4+ CD25+ CD127- Foxp3+ Treg in vitro in the presence of interleukin-2. Moreover, the expanded Treg maintain their phenotype and suppressive activity. These data provide a step toward the use of macaque dendritic cell to expand Treg for future preclinical testing.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células Dendríticas/imunologia , Ativação Linfocitária , Monócitos/citologia , Monócitos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos CD34/imunologia , Imunofenotipagem , Subunidade alfa de Receptor de Interleucina-2/imunologia , Receptores de Lipopolissacarídeos/imunologia , Macaca fascicularis , Modelos Animais
15.
Curr Gene Ther ; 7(5): 334-46, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17979680

RESUMO

Efficient gene transfer has been achieved in several animal models using different vector systems, leading to stable transgene expression. The tight control of this expression is now an important outcome for the field of gene therapy. Such regulation is likely to be required for therapeutic applications and in some instances for safety reasons. For this purpose, several regulatable systems depending on small molecules have been developed. Among these, the tetracycline and the rapamycin dependent systems have been largely used. However, if long-term regulation of the transgene has been obtained in small animal models using these inducible systems, when translational studies were initiated in larger animals, the development of an immune response against proteins involved in transgene regulation were often observed. Such immune response was especially documented when using the TetOn tetracycline regulatable system in nonhuman primates (NHP). Humoral and destructive cellular immune responses against the transactivator involved in this regulation system were documented in a large majority of NHP leading to the complete loss of the transgene regulation and expression. This review will describe the immune responses observed in these different model systems applied for transgene regulation. Focus will be finally given on future directions in which such immune responses might be surmounted, enabling long-term transgene regulation in future clinical developments of gene transfer.


Assuntos
Regulação da Expressão Gênica/imunologia , Vetores Genéticos/administração & dosagem , Regiões Promotoras Genéticas/imunologia , Transgenes/imunologia , Animais , Antibacterianos/farmacologia , Formação de Anticorpos , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/genética , Humanos , Tolerância Imunológica , Imunidade Celular , Imunossupressores/farmacologia , Modelos Animais , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Repressoras/farmacologia , Sirolimo/farmacologia , Tetraciclina/farmacologia , Transativadores/farmacologia , Transgenes/efeitos dos fármacos
16.
Bull Mem Acad R Med Belg ; 161(10-12): 497-508; discussion 508-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17503728

RESUMO

Previous studies have tested gene replacement therapy in RPE65 deficient dogs using recombinant adeno-associated virus 2/2 (rAAV2/2), -2/1 or -2/5 mediated delivery of the RPE65 gene. They all documented restoration of dark- and light-adapted ERG responses and improved psychophysical outcomes. Use of a specific RPE65 promoter and a rAAV vector that targets transgene expression specifically to the retinal pigmented epithelium (RPE) may, however, provide a safer setting for the long-term therapeutic expression of RPE65. Subretinal injection of rAAV2 pseudotyped with serotype 4 (rAAV2/4) specifically targets the RPE. The purpose of our study was to evaluate a rAAV2/4 vector carrying a human RPE65cDNA driven by a human RPE65 promoter, for the ability to restore vision in RPE65-/- purebred Briard dogs. Recombinant rAAV2/4 and rAAV2/2 vectors containing similar human RPE65 promoter and cDNA cassettes were generated and administered subretinally in 8 affected dogs, ages 8 to 30 months (n = 6 with rAAV2/4, n = 2 with rAAV2/2). Although fluorescein angiography and OCT examinations displayed retinal abnormalities in treated retinas, electrophysiological analysis demonstrated that restoration of rod and cone photoreceptor function started as soon as 15 days post-injection, reaching maximal function at 3 months post-injection, and remaining stable thereafter in all animals treated at 8 to 11 months of age. As assessed by the ability of these animals to avoid obstacles in both dim and normal light, functional vision was restored in the treated eye, while the untreated contralateral eye served as an internal control. The dog treated at a later age (30 months) did not recover retinal function or vision, suggesting that there might be a therapeutic window for the successful treatment of RPE65 -/- dogs by gene replacement therapy.


Assuntos
Proteínas de Transporte , Dependovirus/genética , Proteínas do Olho , Terapia Genética , Vetores Genéticos , Atrofia Óptica Hereditária de Leber/terapia , Degeneração Retiniana/terapia , Fatores Etários , Animais , Modelos Animais de Doenças , Cães , Eletrorretinografia , Angiofluoresceinografia , Técnicas de Transferência de Genes , Genes Reporter/genética , Vetores Genéticos/administração & dosagem , Imuno-Histoquímica , Injeções , Epitélio Pigmentado Ocular/metabolismo , Recuperação de Função Fisiológica , Retina/metabolismo , Fatores de Tempo , Resultado do Tratamento , Visão Ocular , cis-trans-Isomerases
17.
Hum Gene Ther Methods ; 27(3): 122-34, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27229628

RESUMO

Recombinant adeno-associated virus (AAV) has emerged as a promising vector for retinal gene delivery to restore visual function in certain forms of inherited retinal dystrophies. Several studies in rodent models have shown that intravitreal injection of the AAV2/2 vector is the optimal route for efficient retinal ganglion cell (RGC) transduction. However, translation of these findings to larger species, including humans, is complicated by anatomical differences in the eye, a key difference being the comparatively smaller volume of the vitreous chamber in rodents. Here, we address the role of the vitreous body as a potential barrier to AAV2/2 diffusion and transduction in the RGCs of dogs and macaques, two of the most relevant preclinical models. We intravitreally administered the AAV2/2 vector carrying the CMV-eGFP reporter cassette in dog and macaque eyes, either directly into the vitreous chamber or after complete vitrectomy, a surgical procedure that removes the vitreous body. Our findings suggest that the vitreous body appears to trap the injected vector, thus impairing the diffusion and transduction of AAV2/2 to inner retinal neurons. We show that vitrectomy before intravitreal vector injection is an effective means of overcoming this physical barrier, improving the transduction of RGCs in dog and macaque retinas. These findings support the use of vitrectomy in clinical trials of intravitreal gene transfer techniques targeting inner retinal neurons.


Assuntos
Terapia Genética , Vetores Genéticos/uso terapêutico , Células Ganglionares da Retina , Animais , Dependovirus/genética , Cães , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde , Humanos , Injeções Intravítreas , Macaca , Retina/patologia , Retina/transplante , Transdução Genética , Vitrectomia
18.
Hum Gene Ther ; 16(9): 1037-46, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16149902

RESUMO

Members of the adeno-associated virus (AAV) family are good candidates for the treatment of ocular diseases because of their relative lack of pathogenicity. We studied the effect of intraocular injection of AAV2-viral IL-10 (vIL-10) on retinal S-antigen-induced experimental autoimmune uveoretinitis (EAU) in Lewis rats. We demonstrated that AAV2/2-GFP injected into the vitreous body transduced the iris and ciliary body, or anterior uvea, and the retina. We showed that intravitreal injection of the AAV2/2-tetON-vIL-10 construct achieved detectable levels of vIL-10 mRNA and protein within the eye and was effective in protecting the rat retina against destruction. This protection was dependent on the level of vIL-10 present in the aqueous humor/ vitreous body. Intravitreal injection of the same construct encased within an AAV5 shell, AAV2/5-tetONvIL- 10, did not confer any degree of protection. It appeared that the AAV2/5 vectors did not transduce the anterior uvea, the site at which inflammatory cells first localize in EAU, nor the ganglion cell layer; induced low expression of vIL-10 mRNA; and did not achieve detectable levels of transgene expression in the aqueous humor/vitreous body. Local treatment with AAV2/2-tetON-vIL-10 did not dampen the systemic immune response, as determined by S-antigen-specific lymphocyte proliferation. Our results show that local intravitreal injection of AAV2/2 is an effective means by which to deliver immunoregulatory molecules into the eye during uveitis, a chronic human ocular disease.


Assuntos
Doenças Autoimunes/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Interleucina-10/uso terapêutico , Retinite/terapia , Uveíte/terapia , Animais , Humor Aquoso/efeitos dos fármacos , Arrestina , Doenças Autoimunes/induzido quimicamente , Doenças Autoimunes/patologia , Proliferação de Células , Dependovirus/genética , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Humanos , Interleucina-10/biossíntese , Interleucina-10/genética , Linfócitos/citologia , Masculino , Ratos , Ratos Endogâmicos Lew , Retina/efeitos dos fármacos , Retina/patologia , Retinite/induzido quimicamente , Retinite/patologia , Tetraciclina/farmacologia , Uveíte/induzido quimicamente , Uveíte/patologia
19.
Curr Gene Ther ; 5(3): 339-48, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15975011

RESUMO

Retinal gene transfer holds big promises for the treatment of inherited and non-inherited blinding diseases, such as retinitis pigmentosa or age-related macular degeneration. Key to the development of successful gene-based therapies for the eye are efficient tools for retinal gene transfer. Vectors based on adeno-associated viruses (AAV) are able to transduce robustly and persistently different retinal cell types of animal models after a single intraocular administration. Recombinant AAV (rAAV) vectors are versatile gene transfer tools in that capsid proteins from dozens of AAV serotypes can be easily interchanged, resulting in the creation of recombinant vectors with unique transduction properties. This has allowed successful proof-of-principle studies using rAAV-mediated gene transfer to restore retinal morphology and function in small and large animal models of retinal diseases. In addition, gene delivery using rAAV vectors in the eye seems to have appropriate biosafety characteristics to rapidly move it from bench to bedside. All the above aspects will be reviewed and discussed in detail below.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos , Doenças Retinianas/terapia , Inibidores da Angiogênese/genética , Inibidores da Angiogênese/uso terapêutico , Animais , Modelos Animais de Doenças , Inativação Gênica , Técnicas de Transferência de Genes , Humanos , Mutação , Doenças Retinianas/genética
20.
Arch Ophthalmol ; 123(4): 500-6, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15824224

RESUMO

OBJECTIVE: To evaluate, in dogs and primates, the short-term effects of subretinal injection and the safety of long-term recombinant adeno-associated virus (rAAV)-mediated transgene expression with respect to retinal morphology and function. METHODS: Subretinal delivery of rAAV (serotype 2, 4, or 5) was performed unilaterally in 14 beagles and 9 macaques. Postsurgical condition was evaluated during a 2-month follow-up study. Three dogs and 1 primate were examined for the long-term study. Green fluorescent protein expression was monitored by fluorescent retinal imaging. Retinal anatomy and function were assessed by angiography and electroretinography, respectively. RESULTS: Transgene expression was observed in 20 of 23 subretinally injected animals (both with and without vitrectomy). We did not detect an inflammatory response in any of the 23 treated subjects. In the long-term study, transgene expression was detected at the latest points evaluated: 36 months for the rAAV-2-injected dog, 24 months for the rAAV-4 and rAAV-5 dogs, and more than 18 months for the rAAV-4-injected primate. Angiography examinations were performed and showed no retinal abnormalities. Functional evaluation showed normal electroretinographic amplitude responses that were similar to those of the noninjected contralateral eyes. CONCLUSIONS: Subretinal injection of the rAAV vector in dogs and primates is a safe procedure with no perioperative complications and a high rate of successful retinal gene transfer. The retinal anatomy and function remained unchanged, despite persistent transgene expression up to 36 months postinjection with rAAV-2, -4, or -5. Additionally, we observed no other adverse effects, such as tumor formation due to possible insertional mutagenesis. These short- and long-term studies on rAAV transgene expression using large animals are encouraging for the prospects of ocular gene therapy applications in humans. CLINICAL RELEVANCE: These short- and long-term studies on rAAV transgene expression using large animals are encouraging for the prospects of ocular gene therapy applications in humans.


Assuntos
Dependovirus/genética , Regulação da Expressão Gênica/fisiologia , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/genética , Retina/metabolismo , Animais , Vírus Defeituosos/genética , Cães , Eletrorretinografia , Angiofluoresceinografia , Seguimentos , Vetores Genéticos , Proteínas de Fluorescência Verde/metabolismo , Macaca fascicularis , Cuidados Pós-Operatórios , Segurança , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA