Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gene Ther ; 21(1): 89-95, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24196086

RESUMEN

For gene therapy to improve lung function in cystic fibrosis (CF) subjects, repeated administration of the gene transfer agent over the lifetime of patients is likely to be necessary. This requirement limits the utility of adenoviral and adeno-associated viral vectors (both previously evaluated in CF gene therapy trials) because of induced adaptive immune responses that render repeated dosing ineffective. For CF gene therapy trials, non-viral vectors are currently the only viable option. We previously showed that the cationic lipid formulation GL67A is the most efficient of several non-viral vectors analysed for airway gene transfer. Here, we assessed the efficacy and safety of administering 12 inhaled doses of GL67A complexed with pGM169, a CpG-free plasmid encoding human CFTR complementary DNA, into mice. We show that repeated administration of pGM169/GL67A to murine lungs is feasible, safe and achieves reproducible, dose-related and persistent gene expression (>140 days after each dose) using an aerosol generated by a clinically relevant nebuliser. This study supports progression into the first non-viral multidose lung trial in CF patients.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Terapia Genética , Vectores Genéticos , Lípidos/administración & dosificación , Lípidos/toxicidad , Pulmón/efectos de los fármacos , Plásmidos , Administración por Inhalación , Animales , Terapia Combinada , Fibrosis Quística/patología , Fibrosis Quística/terapia , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Reproducibilidad de los Resultados
2.
Gene Ther ; 18(10): 996-1005, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21512505

RESUMEN

We use both large and small animal models in our pre-clinical evaluation of gene transfer agents (GTAs) for cystic fibrosis (CF) gene therapy. Here, we report the use of a large animal model to assess three non-viral GTAs: 25 kDa-branched polyethyleneimine (PEI), the cationic liposome (GL67A) and compacted DNA nanoparticle formulated with polyethylene glycol-substituted lysine 30-mer. GTAs complexed with plasmids expressing human cystic fibrosis transmembrane conductance regulator (CFTR) complementary DNA were administered to the sheep lung (n=8 per group) by aerosol. All GTAs gave evidence of gene transfer and expression 1 day after treatment. Vector-derived mRNA was expressed in lung tissues, including epithelial cell-enriched bronchial brushing samples, with median group values reaching 1-10% of endogenous CFTR mRNA levels. GL67A gave the highest levels of expression. Human CFTR protein was detected in small airway epithelial cells in some animals treated with GL67A (two out of eight) and PEI (one out of eight). Bronchoalveolar lavage neutrophilia, lung histology and elevated serum haptoglobin levels indicated that gene delivery was associated with mild local and systemic inflammation. Our conclusion was that GL67A was the best non-viral GTA currently available for aerosol delivery to the sheep lung, led to the selection of GL67A as our lead GTA for clinical trials in CF patients.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/terapia , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Liposomas/administración & dosificación , Nanopartículas/administración & dosificación , Polietileneimina/administración & dosificación , Administración por Inhalación , Animales , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , ADN Complementario/administración & dosificación , ADN Complementario/genética , Humanos , Polietilenglicoles , ARN Mensajero/metabolismo , Ovinos
3.
Gene Ther ; 18(2): 182-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20962870

RESUMEN

We have previously shown that recombinant Sendai virus (SeV) vector, derived from murine parainfluenza virus, is one of the most efficient vectors for airway gene transfer. We have also shown that SeV-mediated transfection on second administration, although reduced by 60% when compared with levels achieved after a single dose, is still high because of the efficient transfection achieved by SeV vector in murine airways. Here, we show that these levels further decrease on subsequent doses. In addition, we validated SeV vector repeat administration in a non-natural host model, the sheep. As part of these studies we first assessed viral stability in a Pari LC Plus nebuliser, a polyethylene catheter (PEC) and the Trudell AeroProbe. We also compared the distribution of gene expression after PEC and Trudell AeroProbe administration and quantified virus shedding after sheep transduction. In addition, we show that bronchial brushings and biopsies, collected in anaesthetized sheep, can be used to assess SeV-mediated gene expression over time. Similar to mice, gene expression in sheep was transient and had returned to baseline values by day 14. In conclusion, the SeV vector should be strongly considered for lung-related applications requiring a single administration of the vector even though it might not be suitable for diseases requiring repeat administration.


Asunto(s)
Expresión Génica , Vectores Genéticos , Virus Sendai/genética , Ovinos/genética , Transducción Genética , Animales , Catéteres , Femenino , Técnicas de Transferencia de Gen/instrumentación , Pulmón , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Retratamiento
5.
Gene Ther ; 15(6): 434-42, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18317498

RESUMEN

Bacteria-mediated gene transfer ('bactofection') has emerged as an alternative approach for genetic vaccination and gene therapy. Here, we assessed bactofection of airway epithelial cells in vitro and in vivo using an attenuated Escherichia coli genetically engineered to invade non-phagocytic cells. Invasive E. coli expressing green fluorescent protein (GFP) under the control of a prokaryotic promoter was efficiently taken up into the cytoplasm of cystic fibrosis tracheal epithelial (CFTE29o-) cells and led to dose-related reporter gene expression. In vivo experiments showed that following nasal instillation the vast majority of GFP-positive bacteria pooled in the alveoli. Further, bactofection was assessed in vivo. Mice receiving 5 x 10(8) E. coli carrying pCIKLux, in which luciferase (lux) expression is under control of the eukaryotic cytomegalovirus (CMV) promoter, showed a significant increase (P<0.01) in lux activity in lung homogenates compared to untransfected mice. Surprisingly, similar level of lux activity was observed for the non-invasive control strain indicating that the eukaryotic CMV promoter might be active in E. coli. Insertion of prokaryotic transcription termination sequences into pCIKLux significantly reduced prokaryotic expression from the CMV promoter allowing bactofection to be detected in vitro and in vivo. However, bacteria-mediated gene transfer leads to a significantly lower lux expression than cationic lipid GL67-mediated gene transfer. In conclusion, although proof-of-principle for lung bactofection has been demonstrated, levels were low and further modification to the bacterial vector, vector administration and the plasmids will be required.


Asunto(s)
Células Epiteliales/microbiología , Escherichia coli/fisiología , Terapia Genética/métodos , Alveolos Pulmonares/microbiología , Animales , Línea Celular , Citomegalovirus/genética , Escherichia coli/genética , Infecciones por Escherichia coli/transmisión , Femenino , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Luciferasas/genética , Enfermedades Pulmonares/microbiología , Ratones , Ratones Noqueados , Viabilidad Microbiana , Organismos Modificados Genéticamente , Plásmidos/administración & dosificación , Regiones Promotoras Genéticas
6.
Nanotechnology ; 19(40): 405102, 2008 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-21832609

RESUMEN

Magnetic nanoparticle-based gene transfection has been shown to be effective in combination with both viral vectors and with non-viral agents. In these systems, therapeutic or reporter genes are attached to magnetic nanoparticles which are then focused to the target site/cells via high-field/high-gradient magnets. The technique has been shown to be efficient and rapid for in vitro transfection and compares well with cationic lipid-based reagents, producing good overall transfection levels with lower doses and shorter transfection times. In spite of its potential advantages (particularly for in vivo targeting), the overall transfection levels do not generally exceed those of other non-viral agents. In order to improve the overall transfection levels while maintaining the advantages inherent in this technique, we have developed a novel, oscillating magnet array system which adds lateral motion to the particle/gene complex in order to promote transfection. Experimental results indicate that the system significantly enhances overall in vitro transfection levels in human airway epithelial cells compared to both static field techniques (p<0.005) and the cationic lipids (p<0.001) tested. In addition, it has the previously demonstrated advantages of magnetofection-rapid transfection times and requiring lower levels of DNA than cationic lipid-based transfection agents. This method shows potential for non-viral gene delivery both in vitro and in vivo.

7.
Gene Ther ; 14(20): 1439-47, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17909539

RESUMEN

Over the last two decades gene therapy has moved from preclinical to clinical studies for many diseases ranging from single gene disorders such as cystic fibrosis and Duchenne muscular dystrophy, to more complex diseases such as cancer and cardiovascular disorders. Gene therapy for severe combined immunodeficiency (SCID) is the most significant success story to date, but progress in many other areas has been significant. We asked 20 leaders in the field succinctly to summarize and comment on clinical gene therapy research in their respective areas of expertise and these are published in two parts in the Progress and Prospect series.


Asunto(s)
Ensayos Clínicos como Asunto , Terapia Genética/tendencias , Enfermedad Coronaria/terapia , Fibrosis Quística/terapia , Oftalmopatías/terapia , Terapia Genética/métodos , Enfermedad Granulomatosa Crónica/terapia , Humanos , Enfermedades por Almacenamiento Lisosomal/terapia , Distrofia Muscular de Duchenne/terapia , Enfermedad de Parkinson/terapia , Enfermedades Vasculares Periféricas/terapia , Inmunodeficiencia Combinada Grave/terapia , Deficiencia de alfa 1-Antitripsina/terapia
8.
Gene Ther ; 14(24): 1688-94, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17898794

RESUMEN

Sendai virus (SeV) vector has been shown to efficiently transduce airway epithelial cells. As a precursor to the potential use of this vector for cystic fibrosis (CF) gene therapy, the correct maturation of the SeV vector-derived CF transmembrane conductance regulator (CFTR) protein was examined using biochemical and functional analyses. We constructed a recombinant SeV vector, based on the fusion (F) gene-deleted non-transmissible SeV vector, carrying the GFP-CFTR gene in which the N terminus of CFTR was fused to green fluorescence protein (GFP). This vector was recovered and propagated to high titers in the packaging cell line. Western blotting using an anti-GFP antibody detected both the fully glycosylated (mature) and the core-glycosylated (immature) proteins, indicating that SeV vector-derived GFP-CFTR was similar to endogenous CFTR. We also confirmed the functional channel activity of GFP-CFTR in an iodide efflux assay. The efficient expression of GFP-CFTR, and its apical surface localization, were observed in both MDCK cells in vitro, and in the nasal epithelium of mice in vivo. We concluded that recombinant SeV vector, a cytoplasmically maintained RNA vector, is able to direct production of a correctly localized, mature form of CFTR, suggesting the value of this vector for studies of CF gene therapy.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Virus Sendai/genética , Transducción Genética/métodos , Animales , Línea Celular , Fibrosis Quística/metabolismo , Expresión Génica , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Cavidad Nasal , Perfusión , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/metabolismo , Mucosa Respiratoria/metabolismo
9.
Gene Ther ; 14(19): 1371-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17597790

RESUMEN

The potential for gene therapy to be an effective treatment for cystic fibrosis has been hampered by the limited gene transfer efficiency of current vectors. We have shown that recombinant Sendai virus (SeV) is highly efficient in mediating gene transfer to differentiated airway epithelial cells, because of its capacity to overcome the intra- and extracellular barriers known to limit gene delivery. Here, we have identified a novel method to allow the cystic fibrosis transmembrane conductance regulator (CFTR) cDNA sequence to be inserted within SeV (SeV-CFTR). Following in vitro transduction with SeV-CFTR, a chloride-selective current was observed using whole-cell and single-channel patch-clamp techniques. SeV-CFTR administration to the nasal epithelium of cystic fibrosis (CF) mice (Cftr(G551D) and Cftr(tm1Unc)TgN(FABPCFTR)#Jaw mice) led to partial correction of the CF chloride transport defect. In addition, when compared to a SeV control vector, a higher degree of inflammation and epithelial damage was found in the nasal epithelium of mice treated with SeV-CFTR. Second-generation transmission-incompetent F-deleted SeV-CFTR led to similar correction of the CF chloride transport defect in vivo as first-generation transmission-competent vectors. Further modifications to the vector or the host may make it easier to translate these studies into clinical trials of cystic fibrosis.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Virus Sendai/genética , Aerosoles , Animales , Cloruros/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/virología , Femenino , Expresión Génica , Ingeniería Genética , Vectores Genéticos/genética , Yoduros/metabolismo , Canales Iónicos/metabolismo , Pulmón , Masculino , Ratones , Ratones Noqueados , Mutación , Técnicas de Placa-Clamp , Transducción Genética/métodos
10.
Gene Ther ; 14(9): 768-74, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17301842

RESUMEN

We have assessed if high-frequency ultrasound (US) can enhance nonviral gene transfer to the mouse lung. Cationic lipid GL67/pDNA, polyethylenimine (PEI)/pDNA and naked plasmid DNA (pDNA) were delivered via intranasal instillation, mixed with Optison microbubbles, and the animals were then exposed to 1 MHz US. Addition of Optison alone significantly reduced the transfection efficiency of all three gene transfer agents. US exposure did not increase GL67/pDNA or PEI/pDNA gene transfer compared to Optison-treated animals. However, it increased naked pDNA transfection efficiency by approximately 15-fold compared to Optison-treated animals, suggesting that despite ultrasound being attenuated by air in the lung, sufficient energy penetrates the tissue to increase gene transfer. US-induced lung haemorrhage, assessed histologically, increased with prolonged US exposure. The left lung was more affected than the right and this was mirrored by a lesser increase in naked pDNA gene transfer, in the left lung. The positive effect of US was dependent on Optison, as in its absence US did not increase naked pDNA transfection efficiency. We have thus established proof of principle that US can increase nonviral gene transfer, in the air-filled murine lung.


Asunto(s)
Albúminas , ADN/administración & dosificación , Fluorocarburos , Terapia Genética/métodos , Pulmón/metabolismo , Transfección/métodos , Ultrasonido , Animales , ADN/genética , Expresión Génica , Luciferasas/genética , Enfermedades Pulmonares/terapia , Masculino , Ratones , Ratones Endogámicos BALB C , Polietileneimina
11.
Gene Ther ; 13(14): 1061-7, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16819538

RESUMEN

Our first review on progress and prospects in cystic fibrosis (CF) gene therapy was published in this series in October 2002. We now summarize the progress made since then and comment on the prospects for CF gene therapy over the next couple of years. Three clinical trials have been carried out, further supporting the proof-of-principle that gene transfer to the airway epithelium is feasible. Developments in viral and non-viral vectors, as well as recent alternative strategies such as gene repair, trans-splicing and stem cell therapy will be reviewed.


Asunto(s)
Fibrosis Quística/terapia , Terapia Genética/métodos , Adenoviridae/genética , Ensayos Clínicos como Asunto , Dependovirus/genética , Predicción , Técnicas de Transferencia de Gen , Terapia Genética/tendencias , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Mucosa Respiratoria/metabolismo , Células Madre/metabolismo , Células Madre/virología
12.
Gene Ther ; 13(23): 1628-38, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16791284

RESUMEN

Topical gene transfer to the airways of cystic fibrosis (CF) patients has been inefficient, partly due to extracellular barriers such as sputum. In an attempt to circumvent these, we assessed whether airway epithelial cells can be transfected by intravenous (i.v.) administration of liposome-complexed or "naked" oligonucleotides (ODNs). The conducting airways are the likely target for CF therapy and are supplied by the bronchial circulation. Consequently, we assessed ODN transfer in the mouse trachea and main bronchi as these are supplied by the bronchial circulation. Liposome-protamine-DNA (LPD) complexes were detected in the bronchial circulation but did not transfect conducting airway epithelial cells, even in the presence of microvascular leakage. In contrast, 'naked' ODNs were delivered to 17% (inter-quartile range (IQR) 10-34%) and 35% (IQR 24-59%) of epithelial cells when injected at 500 microg/animal, without and with microvascular leakage, respectively. Two types of nuclear signal were observed; punctate in cells throughout the airways (3%, IQR 2-6%, and 6%, IQR 4-7%, of cells when delivered without and with microvascular leakage, respectively) and diffuse in a small number of epithelial cells in the proximal trachea. ODNs may be relevant to CF in a variety of ways and these data suggest one way towards implementing their use.


Asunto(s)
Bronquios/irrigación sanguínea , Terapia Genética/métodos , Oligonucleótidos/administración & dosificación , Mucosa Respiratoria/metabolismo , Transfección/métodos , Animales , Transporte Biológico , Bradiquinina/farmacología , Núcleo Celular/metabolismo , Fibrosis Quística/metabolismo , Fibrosis Quística/terapia , Citoplasma/metabolismo , Células Epiteliales/metabolismo , Extravasación de Materiales Terapéuticos y Diagnósticos , Expresión Génica , Histamina/farmacología , Inyecciones Intravenosas , Liposomas , Masculino , Ratones , Ratones Endogámicos BALB C , Microcirculación , Microscopía Fluorescente , FN-kappa B/genética , Factor de Activación Plaquetaria/farmacología
13.
Gene Ther ; 13(21): 1545-52, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16738690

RESUMEN

We have assessed whether magnetic forces (magnetofection) can enhance non-viral gene transfer to the airways. TransMAG(PEI), a superparamagnetic particle was coupled to Lipofectamine 2000 or cationic lipid 67 (GL67)/plasmid DNA (pDNA) liposome complexes. In vitro transfection with these formulations resulted in approximately 300- and 30-fold increase in reporter gene expression, respectively, after exposure to a magnetic field, but only at suboptimal pDNA concentrations. Because GL67 has been formulated for in vivo use, we next assessed TransMAG(PEI) in the murine nasal epithelium in vivo, and compared this to naked pDNA. At the concentrations required for in vivo experiments, precipitation of magnetic complexes was seen. After extensive optimization, addition of non-precipitated magnetic particles resulted in approximately seven- and 90-fold decrease in gene expression for naked pDNA and GL67/pDNA liposome complexes, respectively, compared to non-magnetic particles. Thus, whereas exposure to a magnetic field improved in vitro transfection efficiency, translation to the in vivo setting remains difficult.


Asunto(s)
ADN/farmacología , Terapia Genética/métodos , Magnetismo , Mucosa Respiratoria/metabolismo , Transfección/métodos , Animales , Cationes , Línea Celular Tumoral , Fibrosis Quística/metabolismo , Fibrosis Quística/terapia , Expresión Génica , Ingeniería Genética , Humanos , Lípidos/farmacología , Masculino , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Endogámicos BALB C , Material Particulado
14.
Gene Ther ; 13(5): 449-56, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16319950

RESUMEN

Sendai virus (SeV) is able to transfect airway epithelial cells efficiently in vivo. However, as with other viral vectors, repeated administration leads to reduced gene expression. We have investigated the impact of inducing immunological tolerance to immunodominant T-cell epitopes on gene expression following repeated administration. Immunodominant CD4 and CD8 T-cell peptide epitopes of SeV were administered to C57BL/6 mice intranasally 10 days before the first virus administration with transmission-incompetent F-protein-deleted DeltaF/SeV-GFP. At 21 days after the first virus administration, mice were again transfected with DeltaF/SeV. To avoid interference of anti-GFP antibodies, the second transfection was carried out with DeltaF/SeV-lacZ. At 2 days after the final transfection lung beta-galactosidase expression, T-cell proliferation and antibody responses were measured. A state of 'split tolerance' was achieved with reduced T-cell proliferation, but no impact on antiviral antibody production. There was no enhancement of expression on repeat administration; instead, T-cell tolerance was, paradoxically, associated with a more profound extinction of viral expression. Multiple immune mechanisms operate to eradicate viruses from the lung, and these findings indicate that impeding the adaptive T-cell response to the immunodominant viral epitope is not sufficient to prevent the process.


Asunto(s)
Terapia Genética/métodos , Pulmón/inmunología , Virus Sendai/genética , Linfocitos T/inmunología , Vacunas Virales/administración & dosificación , Animales , Antígenos CD4/inmunología , Antígenos CD8/inmunología , Proliferación Celular , Femenino , Expresión Génica , Ingeniería Genética , Tolerancia Inmunológica/genética , Epítopos Inmunodominantes/inmunología , Ratones , Ratones Endogámicos C57BL , Virus Sendai/inmunología , Vacunas Virales/inmunología
15.
Pediatr Pulmonol ; 39(5): 383-91, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15765543

RESUMEN

Although over 1,000 disease-causing mutations in the CFTR gene have been described, the highly variable disease phenotype in cystic fibrosis (CF) cannot be explained on the basis of this gene alone. Both the environment and other non-CFTR genes are likely to be important. The increased understanding of pathophysiological processes in the CF lung has led to several studies on genes in these pathways, including those involved in host defense, mucin production, and airway responsiveness. Additionally, candidate modifiers of the gastrointestinal manifestations of CF have been explored. One of the major aims of such studies is to produce targets for novel drug developments. This review will summarize the field to date and discuss some of the methodological issues important in the design and interpretation of such studies.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Polimorfismo Genético/genética , Fibrosis Quística/fisiopatología , Variación Genética/genética , Genotipo , Humanos , Fenotipo , Proyectos de Investigación
16.
Gene Ther ; 11 Suppl 1: S43-50, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15454956

RESUMEN

Gene therapy is currently being evaluated for a wide range of acute and chronic lung diseases. The requirement of gene transfer into the individual cell types of the complex lung structure will very much depend on the target disease. Over the last decade, the gene therapy community has recognized that there is not even one vector that is good for all applications, but that the gene transfer agent has to be carefully chosen. Gene therapy is particularly attractive for diseases that currently do not have satisfactory treatment options and probably easier for monogenic disorders than for complex diseases. Cystic fibrosis (CF) fulfills these criteria and is therefore a good candidate for gene therapy-based treatment. This review will focus on CF as an example for lung gene therapy and discuss the progress made in this field over the last couple of years.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Pulmón/metabolismo , Animales , Fibrosis Quística/metabolismo , Humanos , Pulmón/virología , Modelos Animales , Transducción Genética/métodos
17.
Gene Ther ; 11(22): 1659-64, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15284837

RESUMEN

Recombinant Sendai virus (SeV)-mediated gene transfer to differentiated airway epithelial cells has shown to be very efficient, because of its ability to overcome the intra- and extracellular barriers known to limit gene delivery. However, this virus is transmission competent and therefore unlikely to be suitable for use in clinical trials. A nontransmissible, replication-competent recombinant SeV has recently been developed by deleting the envelope Fusion (F) protein gene (SeV/DeltaF). Here we show that SeV/DeltaF is able to mediate beta-galactosidase reporter gene transfer to the respiratory tract of mice in vivo, as well as to human nasal epithelial cells in vitro. Further, in an ex vivo model of differentiated airway epithelium, SeV/DeltaF gene transfer was not importantly inhibited by native mucus. When compared to the transmission-competent SeV in vivo, no difference in gene expression was observed at the time of peak expression. The development of an F-defective nontransmissible SeV, which can still efficiently mediate gene transfer to the airway epithelium, represents the first important step towards the use of a cytoplasmic RNA viral vector in clinical trials of gene therapy.


Asunto(s)
Fibrosis Quística/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Mucosa Nasal/enzimología , Virus Sendai/genética , Transducción Genética/métodos , Adulto , Animales , Células Cultivadas , Femenino , Eliminación de Gen , Expresión Génica , Vectores Genéticos/genética , Humanos , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos BALB C , Moco , Ovinos , Tráquea/enzimología , Proteínas del Envoltorio Viral/genética , beta-Galactosidasa/genética
18.
Gene Ther ; 11(16): 1249-55, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15215885

RESUMEN

The aim of this study was to determine the effects of direct intramyocardial pVEGF165 gene transfer on regional myocardial blood flow in a porcine model of chronic myocardial ischaemia. Pigs underwent placement of an ameroid constrictor around the left circumflex coronary artery. After 3 weeks, animals received direct intramyocardial injections of pVEGF165 (20 x 50 microl at 1 microg/microl, n=11) or a plasmid vector encoding chloramphenicol acetyltransferase (20 x 50 microl at 1 microg/microl, n=11) into a specified target area (TA) of the left lateral wall. At 3 weeks after gene transfer, animals underwent final evaluation including a systematic assessment of regional myocardial blood flow (MBF) under resting and stress conditions. In all, 20 animals (10 per group) reached final studies. There was no change in mean arterial blood pressure or Rentrop collateral score from gene delivery to final studies in either group, nor were there differences between study groups. MBF was significantly higher in the areas adjacent to the TA in the VEGF group under resting (P<0.001) and stress conditions (P<0.05). In addition, pVEGF165 gene transfer abolished flow differences between the adjacent areas and the septum. MBF was not different between study groups in the TA, the anterior wall, or the septum. In conclusion, direct intramyocardial pVEGF165 gene transfer significantly improves myocardial blood flow. However, this effect is limited to the myocardial segments adjacent to the area of gene delivery. These data, therefore, demonstrate a spatial 'delivery-efficacy' mismatch with implications for myocardial gene delivery sites and detection of treatment effects in vivo.


Asunto(s)
Circulación Coronaria/genética , Técnicas de Transferencia de Gen , Isquemia Miocárdica/genética , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Presión Sanguínea/fisiología , Cloranfenicol O-Acetiltransferasa/genética , Enfermedad Crónica , Modelos Animales de Enfermedad , Vectores Genéticos , Frecuencia Cardíaca/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Porcinos , Transcripción Genética
19.
Clin Exp Immunol ; 132(1): 1-8, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12653829

RESUMEN

Gene delivery has the potential to offer effective treatment to patients with life-threatening lung diseases such as cystic fibrosis, alpha1-antitrypsin deficiency and lung cancer. Phase I/II clinical trials have shown that, in principle, gene transfer to the lung is feasible and safe. However, gene expression from both viral and non-viral gene delivery systems has been inefficient. In addition to extra- and intracellular barriers, the host innate and acquired immune system represents a major barrier to successful gene transfer to the lung. Results from studies in experimental animals and clinical trials have shown that inflammatory, antibody and T cell responses can limit transgene expression duration and readministration of the gene transfer vector. We will review here how the development of pharmacological and/or immunological agents can modulate the host immune system and the limitations of these strategies. A better understanding of the immunological barriers which exist in the lung might allow for a more sustained expression of the transgene and importantly help overcome the problem of readministration of viral vectors.


Asunto(s)
Terapia Genética/métodos , Enfermedades Pulmonares/terapia , Pulmón/inmunología , Anticuerpos Bloqueadores/inmunología , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Expresión Génica , Vectores Genéticos/administración & dosificación , Humanos , Inmunosupresores/uso terapéutico , Inflamación , Enfermedades Pulmonares/inmunología , Macrófagos/fisiología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Insuficiencia del Tratamiento
20.
Gene Ther ; 9(20): 1344-50, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12364999

RESUMEN

Since the cloning of the cystic fibrosis gene (CFTR) in 1989, 18 clinical trials have been carried out, including five in the 2 years reviewed here. Most trials demonstrated proof-of-principle for gene transfer to the airway. However, gene transfer efficiency with each of the three gene transfer agents (adenovirus (Ad), adeno-associated virus 2 (AAV2) and cationic liposomes) was low, and most likely insufficient to achieve clinical benefit. Here, we will review the clinical and pre-clinical progress for the last 2 years (2000-2001) and briefly speculate on future prospects for the next 2 in CF gene therapy.


Asunto(s)
Fibrosis Quística/terapia , Terapia Genética/métodos , Animales , Ensayos Clínicos Fase I como Asunto , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células Epiteliales/metabolismo , Expresión Génica , Marcación de Gen , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Humanos , Pulmón , Oligonucleótidos Antisentido/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA