Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Gen Virol ; 93(Pt 12): 2606-2613, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22971818

RESUMEN

Seneca Valley virus (SVV-001) is an oncolytic picornavirus with selective tropism for a subset of human cancers with neuroendocrine differentiation. To characterize further the specificity of SVV-001 and its patterns and kinetics of intratumoral spread, bacterial plasmids encoding a cDNA clone of the full-length wild-type virus and a derivative virus expressing GFP were generated. The full-length cDNA of the SVV-001 RNA genome was cloned into a bacterial plasmid under the control of the T7 core promoter sequence to create an infectious cDNA clone, pNTX-09. A GFP reporter virus cDNA clone, pNTX-11, was then generated by cloning a fusion protein of GFP and the 2A protein from foot-and-mouth disease virus immediately following the native SVV-001 2A sequence. Recombinant GFP-expressing reporter virus, SVV-GFP, was rescued from cells transfected with in vitro RNA transcripts from pNTX-11 and propagated in cell culture. The proliferation kinetics of SVV-001 and SVV-GFP were indistinguishable. The SVV-GFP reporter virus was used to determine that a subpopulation of permissive cells is present in small-cell lung cancer cell lines previously thought to lack permissivity to SVV-001. Finally, it was shown that SVV-GFP administered to tumour-bearing animals homes in to and infects tumours whilst having no detectable tropism for normal mouse tissues at 1×10(11) viral particles kg(-1), a dose equivalent to that administered in ongoing clinical trials. These infectious clones will be of substantial value in further characterizing the biology of this virus and as a backbone for the generation of additional oncolytic derivatives.


Asunto(s)
Virus Oncolíticos/genética , Picornaviridae/genética , Secuencia de Aminoácidos , Animales , Carcinoma de Células Pequeñas/terapia , Carcinoma de Células Pequeñas/virología , Línea Celular Tumoral , Clonación Molecular , Efecto Citopatogénico Viral/genética , ADN Complementario/genética , ADN Viral/genética , Femenino , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Humanos , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/virología , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Viroterapia Oncolítica
2.
J Virol ; 85(9): 4452-61, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21325406

RESUMEN

The RNA genome of Seneca Valley virus (SVV), a recently identified picornavirus, contains an internal ribosome entry site (IRES) element which has structural and functional similarity to that from classical swine fever virus (CSFV) and hepatitis C virus, members of the Flaviviridae. The SVV IRES has an absolute requirement for the presence of a short region of virus-coding sequence to allow it to function either in cells or in rabbit reticulocyte lysate. The IRES activity does not require the translation initiation factor eIF4A or intact eIF4G. The predicted secondary structure indicates that the SVV IRES is more closely related to the CSFV IRES, including the presence of a bipartite IIId domain. Mutagenesis of the SVV IRES, coupled to functional assays, support the core elements of the IRES structure model, but surprisingly, deletion of the conserved IIId(2) domain had no effect on IRES activity, including 40S and eIF3 binding. This is the first example of a picornavirus IRES that is most closely related to the CSFV IRES and suggests the possibility of multiple, independent recombination events between the genomes of the Picornaviridae and Flaviviridae to give rise to similar IRES elements.


Asunto(s)
Picornaviridae/genética , Biosíntesis de Proteínas , ARN Viral/genética , Ribosomas/metabolismo , Animales , Extractos Celulares , Línea Celular , Virus de la Fiebre Porcina Clásica/genética , Análisis Mutacional de ADN , Humanos , Mutación , Conformación de Ácido Nucleico , Pestivirus/genética , Picornaviridae/química , ARN Viral/química , ARN Viral/metabolismo , Conejos , Eliminación de Secuencia
3.
Clin Cancer Res ; 17(4): 888-95, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21304001

RESUMEN

PURPOSE: Seneca Valley Virus (SVV-001) is a novel naturally occurring replication-competent picornavirus with potent and selective tropism for neuroendocrine cancer cell types, including small cell lung cancer. We conducted a first-in-human, first-in-class phase I clinical trial of this agent in patients with cancers with neuroendocrine features, including small cell lung cancer. EXPERIMENTAL DESIGN: Clinical evaluation of single intravenous doses in patients with cancers with neuroendocrine features was performed across five log-increments from 10(7) to 10(11) vp/kg. Toxicity, viral titers and clearance, neutralizing antibody development, and tumor response were assessed. RESULTS: A total of 30 patients were treated with SVV-001, including six with small cell carcinoma at the lowest dose of 10(7) vp/kg. SVV-001 was well tolerated, with no dose-limiting toxicities observed in any dose cohort. Viral clearance was documented in all subjects and correlated temporally with development of antiviral antibodies. Evidence of in vivo intratumoral viral replication was observed among patients with small cell carcinoma, with peak viral titers estimated to be >10(3)-fold higher than the administered dose. One patient with previously progressive chemorefractory small cell lung cancer remained progression-free for 10 months after SVV-001 administration, and is alive over 3 years after treatment. CONCLUSIONS: Intravenous SVV-001 administration in patients is well tolerated at doses up to 10(11) vp/kg, with predictable viral clearance kinetics, intratumoral viral replication, and evidence of antitumor activity in patients with small cell lung cancer. Phase II clinical evaluation in small cell lung cancer is warranted, and has been initiated.


Asunto(s)
Neoplasias Pulmonares/terapia , Viroterapia Oncolítica , Picornaviridae , Carcinoma Pulmonar de Células Pequeñas/terapia , Adulto , Anciano , Femenino , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/virología , Masculino , Persona de Mediana Edad , Carcinoma Pulmonar de Células Pequeñas/inmunología , Carcinoma Pulmonar de Células Pequeñas/patología , Carcinoma Pulmonar de Células Pequeñas/virología , Resultado del Tratamiento , Carga Viral
4.
J Natl Cancer Inst ; 99(21): 1623-33, 2007 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-17971529

RESUMEN

BACKGROUND: Numerous clinical trials have demonstrated that oncolytic viruses can elicit antitumor responses when they are administered directly into localized cancers. However, the treatment of metastatic disease with oncolytic viruses has been challenging due to the inactivation of viruses by components of human blood and/or to inadequate tumor selectivity. METHODS: We determined the cytolytic potential and selectivity of Seneca Valley Virus-001 (SVV-001), a newly discovered native picornavirus, in neuroendocrine and pediatric tumor cell lines and normal cells. Suitability of the virus for intravenous delivery in humans was assessed by blood inactivation assays. Safety was evaluated in vivo using an immune-competent mouse model, and efficacy was evaluated in vivo in athymic mice bearing tumors derived from human small-cell lung cancer and retinoblastoma cell lines. RESULTS: Cell lines derived from small-cell lung cancers and solid pediatric cancers were at least 10,000-fold more sensitive to the cytolytic activity of SVV-001 than were any of the adult normal human cells tested. Viral infectivity was not inhibited by human blood components. Intravenous doses up to 1 x 10(14) virus particles (vp) per kg were well tolerated, and no dose-limiting toxicity was observed in immune-competent mice. A single intravenous dose of 1 x 10(8) vp per kg into athymic mice bearing preestablished small-cell lung or retinoblastoma tumors resulted in complete, durable responses in ten of ten and five of eight mice, respectively. CONCLUSIONS: SVV-001 has potent cytolytic activity and high selectivity for tumor cell lines having neuroendocrine properties versus adult normal cells. Systemically administered SVV-001 has potential for the treatment of metastatic neuroendocrine cancers.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Neuroendocrino/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos , Picornaviridae , Animales , Carcinoma Neuroendocrino/tratamiento farmacológico , Carcinoma de Células Pequeñas/terapia , Línea Celular Tumoral , Modelos Animales de Enfermedad , Pruebas de Hemaglutinación , Humanos , Inmunohistoquímica , Inyecciones Intravenosas , Neoplasias Pulmonares/terapia , Ratones , Ratones Desnudos , Virus Oncolíticos/clasificación , Virus Oncolíticos/patogenicidad , Picornaviridae/clasificación , Picornaviridae/patogenicidad , Proyectos de Investigación , Retinoblastoma/terapia , Trasplante Heterólogo
5.
Methods Mol Med ; 130: 61-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17401164

RESUMEN

One of the most time-consuming steps in the generation of adenoviral vectors is the construction of recombinant plasmids. This chapter describes a detailed method for the rapid construction of adenoviral vectors. The method described here uses homologous recombination machinery of Escherichia coli BJ5183 to construct plasmids used in generation of adenoviral vectors. With this method, no ligation steps are involved in generating the plasmids, and any region of the adenoviral genome can be easily modified. Briefly, the full-length adenoviral genome flanked by unique restriction enzyme sites is first cloned into a bacterial plasmid. Next, the region of the viral genome to be modified is subcloned into a bacterial shuttle plasmid, and the desired changes are introduced by molecular biology techniques. The modified viral DNA fragment is gel-purified and cotransformed with the full-length plasmid, linearized in the targeted region, into BJ5183 cells. Homologous recombination in E. coli generates plasmids containing the modified adenoviral genome. Recombinant virus is generated following release of the viral DNA sequences from the plasmid backbone and transfection into a producer cell line. With this method, homogeneous recombinant adenoviruses can be obtained without plaque purification.


Asunto(s)
Adenoviridae/genética , Escherichia coli/genética , Recombinación Genética , ADN Viral/genética , Vectores Genéticos , Genoma Viral , Indicadores y Reactivos , Plásmidos , Transfección/métodos
6.
Clin Cancer Res ; 12(9): 2869-78, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16675583

RESUMEN

PURPOSE: The purpose of this study was to evaluate a fiber knob replacement strategy to improve infectivity and efficacy of Ad5 fiber chimeric oncolytic viruses for treatment of melanoma and head and neck cancers (HNC). EXPERIMENTAL DESIGN: Adenoviral receptors and transduction levels were used to determine the level of infectivity of fiber-modified, green fluorescent protein-expressing, replication-deficient viruses in a panel of melanoma and HNC cell lines in vitro. Virus yield and cytotoxicity assays were used to determine the tumor specificity and virus replication-mediated cytotoxicity of the fiber-modified oncolytic viruses in the same panel of melanoma and HNC in vitro. Xenograft tumor models were used to assess the antitumor activity of those fiber-modified chimeric viruses compared with the parental virus. RESULTS: Marker gene expression following gene transfer of the fiber chimeric vectors in melanoma and HNC cell lines was approximately 10-fold higher than that obtained with parental Ad5 vector. The fiber chimeric oncolytic variants mediated killing of melanoma and HNC cells that was 2- to 576-fold better than with the parental virus. In addition, fiber chimeric variants produced 2- to 7-fold more progeny virus in tumor cells than the parental virus. Moreover, a high multiplicity of infection was needed for the fiber chimeric viruses to produce cytotoxicity in normal cells. A significantly stronger antitumor response and survival advantage were shown in the tested melanoma and HNC xenograft models following i.t. injections. CONCLUSIONS: In vitro and in vivo studies showed the improved transduction, replication, cytotoxicity, antitumor efficacy, and survival advantage in melanoma and HNC tumor models, suggesting a potential use of these oncolytic agents for the treatment of melanoma and HNCs.


Asunto(s)
Adenovirus Humanos/genética , Técnicas de Transferencia de Gen , Neoplasias de Cabeza y Cuello/terapia , Melanoma/terapia , Línea Celular Tumoral , Quimera , Cartilla de ADN , Marcadores Genéticos , Humanos , Reacción en Cadena de la Polimerasa , Transducción Genética
7.
Clin Cancer Res ; 12(1): 305-13, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16397056

RESUMEN

PURPOSE: The purpose of this study was to examine the tumor specificity, cytotoxicity, and granulocyte macrophage colony-stimulating factor expression of CG0070, a conditionally replicating oncolytic adenovirus, in human bladder transitional cell carcinoma (TCC) cell lines and determine its antitumor efficacy in bladder TCC tumor models. EXPERIMENTAL DESIGN: Virus yield and cytotoxicity assays were used to determine tumor specificity and virus replication-mediated cytotoxicity of CG0070 in a panel of human bladder TCC cell lines and primary cells in vitro. Two s.c. and one orthotopic bladder TCC xenograft tumor models were used to assess antitumor activity of CG0070. RESULTS: In a matched isogenic pair of cell lines with differing retinoblastoma (Rb) pathway status, CG0070 showed selective E1a and granulocyte macrophage colony-stimulating factor (GM-CSF) expression in Rb pathway-defective cells. CG0070 replicated in Rb-defective bladder TCC cell lines as efficiently as wild-type adenovirus but produced 100-fold less virus in normal human cells. CG0070 was up to 1,000-fold more cytotoxic in Rb pathway-defective bladder TCC cells in comparison with normal human cells. Antitumor activity of CG0070 was shown in two bladder TCC s.c. xenograft tumor models following intratumoral injections and intravesical treatment in an orthotopic xenograft tumor model when compared with PBS treatment. CONCLUSIONS: In vitro and in vivo studies showed the selective replication, cytotoxicity, GM-CSF production, and antitumor efficacy of CG0070 in several bladder TCC models, suggesting a potential utility of this oncolytic agent for the treatment of bladder cancer. Further studies are warranted to show the role of human GM-CSF in the antitumor efficacy of CG0070.


Asunto(s)
Carcinoma de Células Transicionales/virología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Viroterapia Oncolítica , Neoplasias de la Vejiga Urinaria/virología , Adenoviridae/genética , Proteínas E1A de Adenovirus/biosíntesis , Animales , Antineoplásicos Fitogénicos/uso terapéutico , Carcinoma de Células Transicionales/terapia , Línea Celular Tumoral , Docetaxel , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Virus Oncolíticos , Proteína de Retinoblastoma/biosíntesis , Proteína de Retinoblastoma/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Taxoides/uso terapéutico , Neoplasias de la Vejiga Urinaria/terapia , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Virus Res ; 116(1-2): 119-28, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16242804

RESUMEN

Out of 51 human adenoviral serotypes recognized to date, 32 of them belong to species D. Members of species D adenoviruses are commonly isolated from immune suppressed patients (organ transplant) and patients suffering from AIDS. The role of species D adenoviruses in pathogenesis is currently unclear. To derive new insights into the genetic content and evolution of species D adenoviruses and as a first step towards development of human adenovirus serotype 46 (Ad46) as vector, the complete nucleotide sequence of the virus was determined. The size of the genome is 35,178 bp in length with a G+C content of 56.9%. All the early and late region genes are present in the expected locations of the genome. The deduced amino acid sequences of all late region genes, with the exception of fiber, exhibited high degree of homology with the corresponding proteins of other adenoviruses. The deduced amino acid sequences of early regions E1, E3 and E4 showed a high degree of homology with the corresponding proteins of adenoviruses belonging to species D and less homology with the corresponding proteins of adenoviruses of other species. The homologues of Ad5 E3 region genes encoding 12.5K, gp19K, 10.4K, 14.5K and 14.7K are conserved in the genome of Ad46. However, the E3 region of Ad46 lacks genes encoding 6.7K and adenovirus death protein (ADP) but contains two additional open reading frames with a coding capacity of 433 and 281 amino acids. The fiber protein of Ad46 is 200 amino acids smaller than the fiber protein of Ad5 and contains only 10 pseudo-repeats in the shaft region. To facilitate the manipulation of the genome, the complete genome of Ad46 was cloned into a single bacterial plasmid. Following transfection into E1 complementing cell lines, the virus was recovered demonstrating the feasibility of viral genome manipulation for generation of recombinant viruses.


Asunto(s)
Adenovirus Humanos/genética , Genoma Viral , Proteínas Precoces de Adenovirus/genética , Composición de Base , Secuencia de Bases , Línea Celular , Secuencia Conservada , Orden Génico , Humanos , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Filogenia , ARN Viral , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Serotipificación , Proteínas Virales/genética
9.
Cancer Gene Ther ; 11(8): 555-69, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15232601

RESUMEN

A potentially promising treatment of metastatic cancer is the systemic delivery of oncolytic adenoviruses. This requires engineering viruses which selectively replicate in tumors. We have constructed such an oncolytic adenovirus, OAS403, in which two early region genes are under the control of tumor-selective promoters that play a role in two key pathways involved in tumorigenesis. The early region E1A is controlled by the promoter for the E2F-1 gene, a transcription factor that primarily upregulates genes for cell growth. The E4 region is under control of the promoter for human telomerase reverse transcriptase, a gene upregulated in most cancer cells. OAS403 was evaluated in vitro on a panel of human cells and found to elicit tumor-selective cell killing. Also, OAS403 was less toxic in human hepatocyte cultures, as well as in vivo when compared to an oncolytic virus that lacked selective E4 control. A single intravenous injection of 3 x 10(12) vp/kg in a Hep3B xenograft mouse tumor model led to significant antitumor efficacy. Additionally, systemic administration in a pre-established LNCaP prostate tumor model resulted in over 80% complete tumor regressions at a tolerable dose. Vector genome copy number was measured in tumors and livers at various times following tail vein injection and showed a selective time-dependent increase in tumors but not livers over 29 days. Furthermore, efficacy was significantly improved when OAS403 treatment was combined with doxorubicin. This virus holds promise for the treatment of a broad range of human cancers including metastatic disease.


Asunto(s)
Adenoviridae/genética , Neoplasias/terapia , Adenoviridae/metabolismo , Animales , Proteínas de Unión al ADN , Doxorrubicina/uso terapéutico , Vectores Genéticos/administración & dosificación , Hepatocitos/metabolismo , Humanos , Concentración 50 Inhibidora , Inyecciones , Ratones , Ratones SCID , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/metabolismo , Regiones Promotoras Genéticas , Telomerasa/genética , Telomerasa/metabolismo , Replicación Viral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Cell Physiol ; 199(3): 399-411, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15095287

RESUMEN

Vascular endothelial growth factor (VEGF) and insulin-like growth factor-1 (IGF-1) have been implicated as important stimulatory factors for retinal neovascularization. In this study, we used intraocular gene transfer with gutless adenoviral (AGV) vectors to determine the effect of increased intraocular expression of VEGF, IGF-1, or sphingosine kinase (SPK), which produces sphingosine-1-phosphate, another angiogenic factor. Retinal neovascularization did not occur from intravitreous AGV-vectored VEGF, IGF-1, SPK, or combined VEGF and IGF-1, except occasionally adjacent to the retinal penetration site from the injection. However, corneal and iris neovascularization occurred after 2 weeks in all eyes injected with AGV.VEGF, but not those injected with only AGV.IGF-1 or AGV.SPK. These data suggest that the superficial capillary bed of the retina is relatively insensitive to VEGF, IGF-1, or SPK in adult mice, except when combined with retinal trauma. However, AGV-vectored VEGF is sufficient to consistently cause severe corneal and iris neovascularization. This provides a model for anterior segment neovascularization, which unlike previous models is relatively inexpensive and is not plagued by spontaneous regression, and therefore, may be useful for identification of new treatments.


Asunto(s)
Adenoviridae , Ojo/irrigación sanguínea , Neovascularización Retiniana , Transducción Genética , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Adenoviridae/genética , Animales , Cartilla de ADN , Vectores Genéticos , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Factor I del Crecimiento Similar a la Insulina/genética , Operón Lac , Lisofosfolípidos/metabolismo , Ratones , Neovascularización Fisiológica/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/biosíntesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
11.
J Cell Physiol ; 199(3): 412-7, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15095288

RESUMEN

Increased expression of vascular endothelial growth factor (VEGF) in the retina starting after postnatal day (P)7 results in neovascularization originating from deep retinal capillaries, but not those in the superficial capillary bed. Doxycycline was administered starting P0 to double transgenic mice with inducible expression of VEGF in the retina. These mice showed proliferation and dilation of superficial retinal capillaries, indicating that at this stage of development, the superficial capillaries are sensitive to the effects of VEGF. Angiopoietin-2 (Ang2) is expressed along the surface of the retina for several days after birth, but by P7 and later, Ang2 is only expressed in the region of the deep capillary bed. In mice with ubiquitous doxycycline-inducible expression of Ang2, in the absence of doxycycline, intravitreous injection of a gutless adenoviral vector expressing VEGF (AGV.VEGF) resulted in neovascularization of the cornea and iris, but no retinal neovascularization. After treatment with doxycycline to induce Ang2 expression, intravitreous injection of AGV.VEGF caused retinal neovascularization in addition to corneal and iris neovascularization. The retinal neovascularization originated from both the superficial and deep capillary beds. These data suggest that Ang2 promotes sensitivity to the angiogenic effects of VEGF in retinal vessels.


Asunto(s)
Angiopoyetina 2/farmacología , Neovascularización Fisiológica , Retina/crecimiento & desarrollo , Neovascularización Retiniana , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Adenoviridae/genética , Animales , Animales Recién Nacidos , Antibacterianos/farmacología , Doxiciclina/farmacología , Vectores Genéticos , Ratones , Ratones Transgénicos , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/farmacología
12.
Hum Gene Ther ; 14(8): 749-61, 2003 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-12804138

RESUMEN

Transcriptional regulation that is rapid, reversible, and repeatedly inducible would greatly enhance the safety and efficacy of many gene therapy strategies. We developed a chimeric ligand-inducible regulation system based on the human estrogen receptor. This system has two components, the responsive promoter driving expression of the transgene of interest, and the ligand-inducible chimeric transcription factor. The transcription factor is composed of a novel DNA binding domain and a modified estrogen receptor ligand-binding domain. A point mutation in the ligand-binding domain significantly reduces estrogen binding while allowing binding of the estrogen antagonist, tamoxifen. We used a gutless adenoviral vector system and incorporated both components into two separate vectors. A single gutless vector encoding both system components was also generated. The tamoxifen-mediated induciblity of transgene expression of the gutless vector system was compared in vitro and in vivo with the analogous components incorporated into early generation, E1/E2a/E3-deficient adenoviral vectors. In normal mice, both the gutless vector and early generation systems displayed inducibility in the presence of tamoxifen. Importantly, the gutless vector system was inducible to extremely high levels, at least four times over a 2-month period. In contrast, the early generation vector system was inducible only once. Furthermore, the early generation system displayed significant toxicity, as evidenced by extremely high liver enzyme levels, abnormal liver pathology, and rapid loss of vector DNA from the liver, while the gutless vector system displayed minimal toxicity. These data directly demonstrate the improved in vivo function of the tamoxifen-inducible transcriptional regulation system in the context of the gutless adenoviral vectors.


Asunto(s)
Adenoviridae/genética , Regulación de la Expresión Génica , Vectores Genéticos , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas , Virus Defectuosos/genética , Endostatinas/biosíntesis , Endostatinas/genética , Vectores Genéticos/toxicidad , Células HeLa , Humanos , Ligandos , Hígado/metabolismo , Hígado/patología , Hepatopatías/metabolismo , Hepatopatías/patología , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología , Transcripción Genética
13.
Hum Gene Ther ; 14(3): 243-54, 2003 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-12639304

RESUMEN

Adenoviral vectors devoid of all viral coding regions are referred to by many names, including gutless vectors. Gutless vectors display reduced toxicity and immunogenicity, increased duration of transgene expression, and increased coding capacity compared to early generation vectors, which contain the majority of the viral backbone genes. However, the production of gutless vectors at a scale and purity suitable for clinical use has limited the utility of this technology. In this work we describe the optimization of the production of gutless vectors. We constructed an improved helper virus and generated an alternative gutless vector producer cell line, PERC6-Cre. We demonstrated increased gutless vector yields, minimal helper virus contamination, and no replication-competent adenovirus contamination using the optimized system. Furthermore, the PERC6-Cre cells were adapted to serum-free suspension culture and high-titer gutless vector preparations were produced using bioreactor technology, suggesting the feasibility of gutless vector scale-up for clinical use. Finally, we observed that helper virus lacking a packaging signal could be packaged at a low frequency, revealing an inherent limitation to the differential packaging strategy for gutless vector propagation.


Asunto(s)
Adenoviridae , Vectores Genéticos , Virus Helper , Genes Reporteros , Reacción en Cadena de la Polimerasa
14.
Mol Ther ; 5(1): 63-73, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11786047

RESUMEN

Gutless adenoviral vectors are devoid of all viral coding regions and display reduced cytotoxicity, diminished immunogenicity, and an increased coding capacity compared with early generation vectors. Using hemophilia A, a deficiency in clotting factor VIII (FVIII), as a model disease, we generated and evaluated a gutless vector encoding human FVIII. The FVIII gutless vector grew to high titer and was reproducibly scaled-up from vector seed lots. Extensive viral DNA analyses revealed no rearrangements of the vector genome. A quantitative PCR assay demonstrated helper virus contamination levels of <2%, with the best preparation containing 0.3% helper virus. We compared the gutless vector with an E1/E2a/E3-deficient (Av3) early generation vector encoding an identical FVIII expression cassette following intravenous administration to hemophilia A mice. Gutless vector-treated mice displayed 10-fold higher FVIII expression levels that were sustained for at least 9 months. In contrast, mice treated with the Av3 vector displayed FVIII levels below the limit of sensitivity of the assay at 3 months. Assessment of hepatotoxicity by measuring the serum levels of liver enzymes demonstrated that the gutless vector was significantly less toxic than the Av3 vector at time points later than 7 days. At the highest dose used, both vectors caused a transient 10-fold increase in liver enzymes 1 day after vector administration, suggesting that this increase was caused by direct toxicity of the input capsid proteins. These data demonstrate that the gutless vector displayed increased duration and levels of FVIII expression, and was significantly less toxic than an analogous early generation vector.


Asunto(s)
Adenoviridae/genética , Factor VIII/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Hemofilia A/genética , Animales , Secuencia de Bases , Cartilla de ADN , Factor VIII/administración & dosificación , Femenino , Terapia Genética , Hemofilia A/terapia , Humanos , Ratones , Reacción en Cadena de la Polimerasa , Transducción Genética
15.
J Gen Virol ; 80 ( Pt 11): 2909-2916, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10580052

RESUMEN

Porcine adenovirus has been proposed as a potential vector for generating novel and effective vaccines for pigs. As a prerequisite for the generation of helper-dependent porcine adenovirus-3 (PAV-3) vectors, two E1-complementing porcine cell lines expressing E1 proteins of human adenovirus-5 (HAV-5) were made. These cell lines could be efficiently transfected with DNA and allowed the rescue and propagation of a PAV-3 recombinant, PAV201, containing a 0.597 kb E3 deletion and a 0.803 kb E1A deletion. Our data demonstrate that E1A proteins of HAV-5 have the capacity to transform foetal porcine retina cells and complement for the E1A proteins of PAV-3. The green fluorescent protein (GFP) gene placed under the control of a cytomegalovirus immediate early promoter was inserted into the E1A region of the PAV201 genome. Using these cell lines, a helper-dependent PAV-3 recombinant expressing GFP, PAV202, was constructed and characterized. The wild-type PAV-3 and the recombinant PAV202 expressing GFP were used to determine the ability of the virus to enter and replicate in cells of human and animal origin under cell culture conditions. Our results suggest that PAV-3 enters but does not replicate in dog, sheep, bovine and human cells.


Asunto(s)
Virus Defectuosos/genética , Vectores Genéticos , Mastadenovirus/genética , Porcinos/virología , Replicación Viral , Proteínas E1 de Adenovirus/fisiología , Animales , Bovinos , Línea Celular , Perros , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C3H
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...