Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cytotherapy ; 10(5): 526-39, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18821360

RESUMEN

BACKGROUND: Retroviral vectors are regularly used to transduce stem cells and their derivatives for experimental and therapeutic purposes. Because these vectors integrate semi-randomly into the cellular genome, analysis of integranated retroviral DNA/host cell DNA junctions (IHJ) facilitates clonality studies of engrafted cells, allowing their differentiation, survival and fate to be tracked. In the case of any adverse events, IHJ analysis can allow the identification of potentially oncogenic integration sites. At present, most measures to assess IHJ are complex, insensitive and may be subject to IHJ selection bias inherent to the technology used. METHODS: We have developed and validated a simple but effective technique for generating libraries of IHJ, which we term flanking-sequence exponential anchored-polymerase chain reaction (FLEA-PCR). Flanking-sequence random anchoring is used as an alternative to restriction enzyme digestion and cassette ligation to allow consistent detection of IHJ and decrease bias. RESULTS: Individual clones from plasmid libraries can be sequenced and assembled using custom-written software, and FLEA-PCR smears can be analyzed by capillary electrophoresis after digestion with restriction enzymes. DISCUSSION: This approach can readily analyze complex mixtures of IHJ, allowing localization of these sequences to their genomic sites. This approach should simplify analysis of retroviral integration.


Asunto(s)
ADN Cruciforme/análisis , ADN Viral/análisis , ADN/análisis , Interacciones Huésped-Patógeno , Reacción en Cadena de la Polimerasa/métodos , Retroviridae/genética , Integración Viral/genética , Separación Celular , Citometría de Flujo , Vectores Genéticos , Células HeLa , Humanos , Leucocitos Mononucleares , Reacción en Cadena de la Polimerasa/instrumentación , Secuencias Repetidas Terminales/genética , Transducción Genética
2.
Clin Cancer Res ; 7(9): 2870-9, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11555605

RESUMEN

Inhibition of tumor-induced neovascularization appears to be an effective anticancer approach, although long-term angiogenesis inhibition may be required. An alternative to chronic drug administration is a gene therapy-mediated approach in which long-term in vivo protein expression is established. We have tested this approach by modifying murine bone marrow-derived cells with a gene encoding an angiogenesis inhibitor: a soluble, truncated form of the vascular endothelial growth factor receptor-2, fetal liver kinase-1 (Flk-1). Murine bone marrow cells were transduced with a retroviral vector encoding either truncated, soluble Flk-1 (tsFlk-1) together with green fluorescent protein (GFP) or GFP alone. Tumor growth in mice challenged 3 months after transplantation with tsFlk-1-expressing bone marrow cells was significantly inhibited when compared with tumor growth in control-transplanted mice. Immunohistochemical analysis of tumors in each group demonstrated colocalization of GFP expression in cells staining with endothelial cell markers, suggesting that the endothelial cells of the tumor-induced neovasculature were derived, at least in part, from bone marrow precursors. These results suggest that long-term expression of a functional angiogenesis inhibitor can be generated through gene-modified, bone marrow-derived stem cells, and that this approach can have significant anticancer efficacy. Modifying these cells seems to have the added potential benefit of targeting transgene expression to the tumor neovasculature, because bone marrow-derived endothelial cell precursors seem to be recruited in the process of tumor-induced angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/genética , Células de la Médula Ósea/metabolismo , Neoplasias Experimentales/prevención & control , Neovascularización Patológica/prevención & control , Inhibidores de la Angiogénesis/metabolismo , Animales , División Celular/genética , Femenino , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes , Trasplante de Células Madre Hematopoyéticas , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos , Ratones SCID , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular , Transfección , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Gene Ther ; 8(7): 537-45, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11498776

RESUMEN

Modalities that act through different mechanisms can often provide synergistic antitumor activity for the treatment of refractory tumors when used in combination. Here we report a gene therapy approach in which the genes for the angiogenesis inhibitor, endostatin, and the marker protein and potent immunogen, green fluorescent protein (GFP), were delivered to murine neuroblastoma cells prior to inoculation of the tumor cells into syngeneic immunocompetent mice. Although the effect of either angiogenesis inhibition or immunomodulation alone resulted in only a modest delay in tumor growth, when these approaches were used in combination, prevention of the formation of appreciable tumors was effected in 15 of 24 (63%) mice. The combination of endostatin and GFP expression elicited a strong immune response that was T cell-mediated and was reactive against both GFP and tumor cell line-specific antigens. This afforded treated mice protection against subsequent tumor challenge with unmodified tumor cells. These results suggest that antiangiogenic and immunotherapy strategies, when used in a gene therapy-mediated approach, can act synergistically in an effective multimodality anticancer approach.


Asunto(s)
Colágeno/biosíntesis , Colágeno/genética , Terapia Genética/métodos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Neuroblastoma/terapia , Fragmentos de Péptidos/biosíntesis , Fragmentos de Péptidos/genética , Inhibidores de la Angiogénesis/farmacología , Animales , División Celular , Movimiento Celular , Separación Celular , Células Cultivadas , Clonación Molecular , Terapia Combinada , Endostatinas , Endotelio Vascular/citología , Citometría de Flujo , Proteínas Fluorescentes Verdes , Humanos , Inmunoterapia/métodos , Ratones , Ratones SCID , Plásmidos/metabolismo , Biosíntesis de Proteínas , Proteínas Recombinantes/metabolismo , Retroviridae/genética , Linfocitos T/metabolismo , Factores de Tiempo , Transcripción Genética , Transducción Genética , Células Tumorales Cultivadas , Venas Umbilicales/citología
4.
Cancer Res ; 61(15): 5810-6, 2001 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-11479220

RESUMEN

Thioguanine and mercaptopurine are prodrugs requiring conversion into thiopurine nucleotides to exert cytotoxicity. Thiopurine S-methyltransferase (TPMT), an enzyme subject to genetic polymorphism, catabolizes thiopurines into inactive methylated bases, but also produces methylthioguanine nucleotides and methylmercaptopurine nucleotides from thioguanine and mercaptopurine nucleotides, respectively. To study the effect of TPMT on activation versus inactivation of mercaptopurine and thioguanine, we used a retroviral gene transfer technique to develop human CCRF-CEM cell lines that did (TPMT+) and did not (MOCK) overexpress TPMT. After transduction, TPMT activities were 14-fold higher in the TPMT+ versus the MOCK cell lines (P < 0.001). TPMT+ cells were less sensitive to thioguanine than MOCK cells (IC(50) = 1.10+/- 0.12 microM versus 0.55 +/- 0.19 microM; P = 0.02); in contrast, TPMT+ cells were more sensitive to mercaptopurine than MOCK cells (IC(50) = 0.52 +/- 0.20 microM versus 1.50 +/- 0.23 microM; P < 0.01). The lower sensitivity of TPMT+ versus MOCK cells to thioguanine was associated with lower thioguanine nucleotide concentrations (917 +/- 282 versus 1515 +/- 183 pmol/5 x 10(6) cells; P = 0.01), higher methylthioguanine nucleotide concentrations (252 +/- 34 versus 27 +/- 10 pmol/5 x 10(6) cells; P = 0.01), less inhibition of de novo purine synthesis (13 versus 95%; P < 0.01), and lower deoxythioguanosine incorporation into DNA (2.0 +/- 0.6% versus 7.2 +/- 2.0%; P < 0.001). The higher sensitivity of TPMT+ cells to mercaptopurine was associated with higher concentrations of methylmercaptopurine nucleotide (2601 +/- 1055 versus 174 +/- 77 pmol/5 x 10(6) cells; P = 0.01) and greater inhibition of de novo purine synthesis (>99% versus 74%; P < 0.01) compared with MOCK cells. We conclude that methylation of mercaptopurine contributes to the antiproliferative properties of the drug, probably through inhibition of de novo purine synthesis by methylmercaptopurine nucleotides, whereas thioguanine is inactivated primarily by TPMT.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Leucemia-Linfoma de Células T del Adulto/enzimología , Mercaptopurina/análogos & derivados , Mercaptopurina/farmacología , Metiltransferasas/metabolismo , Tioguanina/farmacología , Células 3T3 , Animales , Antimetabolitos Antineoplásicos/farmacocinética , Biotransformación , Citosol/metabolismo , ADN de Neoplasias/metabolismo , Desoxiguanosina/metabolismo , Técnicas de Transferencia de Gen , Células HeLa , Humanos , Leucemia-Linfoma de Células T del Adulto/genética , Mercaptopurina/farmacocinética , Metiltransferasas/biosíntesis , Metiltransferasas/genética , Ratones , Nucleótidos de Purina/metabolismo , Purinas/biosíntesis , Retroviridae/genética , Tioguanina/farmacocinética , Tionucleósidos/metabolismo , Tionucleótidos/metabolismo , Células Tumorales Cultivadas
5.
Ann N Y Acad Sci ; 938: 262-76; discussion 276-7, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11458516

RESUMEN

Limited functional expression of the viral envelope receptor is a recognized barrier to efficient oncoretroviral mediated gene transfer. To circumvent this barrier we evaluated a number of envelope proteins with respect to gene transfer efficiency into primitive human hematopoietic stem cell populations. We observed that oncoretroviral vectors pseudotyped with the envelope protein of feline endogenous virus (RD114) could efficiently transduce human repopulating cells capable of establishing multilineage hematopoiesis in immunodeficient mice after a single exposure to RD114-pseudotyped vector. Comparable rates of gene transfer with amphotropic and GALV-pseudotyped vectors have been reported, but only after multiple exposures to the viral supernatant. Oncoretroviral vectors pseudotyped with the RD114 or the amphotropic envelopes had similar stability in vitro, indicating that the increased efficiency in gene transfer is at the receptor level likely due to increased receptor expression or an increased receptor affinity for the RD114 envelope. We also found that RD114-pseudotype vectors can be efficiently concentrated, thereby removing any adverse effects of the conditioned media to the long-term repopulating potential of the target human hematopoietic stem cell. These studies demonstrate the potential of RD114-pseudotyped vectors for clinical use.


Asunto(s)
Retrovirus Endógenos/genética , Productos del Gen env/fisiología , Vectores Genéticos , Células Madre Hematopoyéticas/virología , Transfección/métodos , Animales , Células Cultivadas/trasplante , Células Cultivadas/virología , Medios de Cultivo Condicionados/farmacología , Medio de Cultivo Libre de Suero , Resistencia a Medicamentos/genética , Sangre Fetal/citología , Genes Reporteros , Vectores Genéticos/química , Vectores Genéticos/genética , Vectores Genéticos/aislamiento & purificación , Vectores Genéticos/ultraestructura , Proteínas Fluorescentes Verdes , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Humanos , Virus de la Leucemia del Gibón/genética , Virus de la Leucemia Murina/genética , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Recombinantes de Fusión/biosíntesis , Selección Genética , Tetrahidrofolato Deshidrogenasa/biosíntesis , Tetrahidrofolato Deshidrogenasa/genética , Trimetrexato/farmacología , Ultracentrifugación
6.
Mol Ther ; 3(6): 911-9, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11407905

RESUMEN

The hematopoietic stem cell has long been considered an ideal target for the introduction of therapeutic genes to treat human disorders such as Fanconi anemia (FA). Although recent progress in large animal models is encouraging, application to nonmalignant conditions is limited by the perceived necessity of myeloablative conditioning. We and others have shown that very low irradiation doses are sufficient to allow significant hematopoietic engraftment in murine hosts even after the introduction of xenogeneic genes. To determine the degree of engraftment of genetically modified cells attainable with very low irradiation doses in larger animals, we employed the rhesus macaque competitive repopulation model. Four animals underwent mobilization with stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) followed by apheresis. The apheresis product was enriched for the CD34-positive fraction by immunomagnetic selection and split equally for transduction with either G1FC26, a retroviral vector carrying the Fanconi anemia complementation group C gene, or PLII, a nonexpression control retroviral vector carrying both neomycin and beta-galactosidase gene sequences modified to prevent translation. Transductions were performed daily in the presence of fresh IL-3, IL-6, SCF, and Flt-3 ligand on fibronectin-coated plates over 96 h. Animals were conditioned with a single dose of either 100 (n = 2) or 200 (n = 2) cGy and received the combined products of transduction on the following day. None of the animals experienced clinically significant neutropenia nor required the use of central line placement, transfusional support with blood products, or intravenous antibiotics. Using real-time PCR, circulating levels of genetically modified cells as high as 1% were initially detected. Stable, albeit, significantly lower levels from both vector-transduced aliquots (<0.1%) persisted beyond 12 months posttransplant in all four animals. Although not sufficient to correct the phenotype in many human disorders, stable low-level engraftment by genetically modified cells following low-intensity conditioning may prove adequate in disorders such as FA due to the selective advantage conferred upon corrected cells.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/efectos de la radiación , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de la radiación , Macaca mulatta/sangre , Proteínas Nucleares , Proteínas/genética , Retroviridae/genética , Acondicionamiento Pretrasplante , Animales , Antígenos CD34/metabolismo , Ensayo de Unidades Formadoras de Colonias , Cartilla de ADN/química , Proteína del Grupo de Complementación C de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Técnicas de Transferencia de Gen , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/virología , Interleucina-3/farmacología , Interleucina-6/farmacología , Proteínas de la Membrana/farmacología , Reacción en Cadena de la Polimerasa , Protectores contra Radiación/farmacología , Transducción Genética , Irradiación Corporal Total
7.
Blood ; 97(5): 1258-65, 2001 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11222368

RESUMEN

Long-term expression of coagulation factor IX (FIX) has been observed in murine and canine models following administration of recombinant adeno-associated viral (rAAV) vectors into either the portal vein or muscle. These studies were designed to evaluate factors that influence rAAV-mediated FIX expression. Stable and persistent human FIX (hFIX) expression (> 22 weeks) was observed from 4 vectors after injection into the portal circulation of immunodeficient mice. The level of expression was dependent on promoter with the highest expression, 10% of physiologic levels, observed with a vector containing the cytomegalovirus (CMV) enhancer/beta-actin promoter complex (CAGG). The kinetics of expression after injection of vector particles into muscle, tail vein, or portal vein were similar with hFIX detectable at 2 weeks and reaching a plateau by 8 weeks. For a given dose, intraportal administration of rAAV CAGG-FIX resulted in a 1.5-fold or 4-fold higher level of hFIX compared to tail vein or intramuscular injections, respectively. Polymerase chain reaction analysis demonstrated predominant localization of the rAAV FIX genome in liver and spleen after tail vein injection with a higher proportion in liver after portal vein injection. Therapeutic levels of hFIX were detected in the majority of immunocompetent mice (21 of 22) following intravenous administration of rAAV vector without the development of anti-hFIX antibodies, but hFIX was not detected in 14 immunocompetent mice following intramuscular administration, irrespective of strain. Instead, neutralizing anti-hFIX antibodies were detected in all the mice. These observations may have important implications for hemophilia B gene therapy with rAAV vectors.


Asunto(s)
Factor IX/genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/farmacocinética , Transducción Genética/métodos , Animales , Anticuerpos/análisis , Anticuerpos/sangre , ADN Complementario/farmacocinética , ADN Recombinante , Dependovirus/genética , Factor IX/inmunología , Humanos , Inyecciones Intramusculares , Inyecciones Intravenosas , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Vena Porta , Factores de Tiempo , Distribución Tisular
8.
Mol Ther ; 3(1): 78-87, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11162314

RESUMEN

Transfer of drug resistance genes to hematopoietic stem cells offers the potential to protect cancer patients from drug-induced myelosuppression and to increase the number of gene-modified cells by in vivo selection. In this study, a retroviral vector expressing both a P140K variant of human O6-methylguanine-DNA methyltransferase (MGMT) and an EGFP reporter gene was evaluated for stem cell protection in a murine transplant model. Mice transplanted with vector-transduced cells showed significant resistance to the myelosuppressive effects of temozolomide (TMZ), an orally administered DNA-methylating drug, and O6-benzylguanine (BG), a drug that depletes cells of wild-type MGMT activity. Following drug treatment, increases in EGFP(+) peripheral blood cells were seen in all peripheral blood lineages, and secondary transplant experiments proved that selection had occurred at the stem cell level. In a second set of experiments in which transduced cells were diluted with unmarked cells, efficient stem cell selection was noted together with progressive marrow protection with repeated treatment courses. Altogether, these results show that P140K MGMT gene transfer can protect stem cells against the toxic effects of TMZ and BG and that this vector/drug system may be useful for clinical myeloprotection and for in vivo selection of transduced stem cells.


Asunto(s)
Transferasas Alquil y Aril/genética , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Vectores Genéticos , Guanina/análogos & derivados , Guanina/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Retroviridae/genética , Animales , Antineoplásicos Alquilantes/farmacología , Linfocitos B/metabolismo , Plaquetas/metabolismo , Southern Blotting , Técnicas de Cultivo de Célula/métodos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Eritrocitos/metabolismo , Femenino , Citometría de Flujo , Granulocitos/metabolismo , Proteínas Fluorescentes Verdes , Trasplante de Células Madre Hematopoyéticas , Hemoglobinas/metabolismo , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Neutrófilos/metabolismo , Plásmidos/metabolismo , Linfocitos T/metabolismo , Temozolomida , Factores de Tiempo
9.
J Pediatr Surg ; 36(1): 30-6, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11150434

RESUMEN

BACKGROUND/PURPOSE: Preventing tumors from forming new blood vessels appears to be an effective new anticancer approach. Antiangiogenic therapy usually is cytostatic, however, and, therefore, long-term angiogenesis inhibition is likely to be required. The objective of this study was to determine if sustained gene therapy-mediated expression of these agents from tumor cells could restrict tumor growth in vivo. METHODS: Two replication-defective retroviral vectors were made, one encoding both the soluble, truncated vascular endothelial growth factor receptor (VEGF-R2), flk-1, together with green fluorescent protein (GFP), and the other encoding GFP alone. These vectors were then used to transduce murine neuroblastoma cells (NXS2). Stable, high expression of the flk-1 transgene was confirmed in the former population of cells by Western analysis. Flk-1 protein was isolated from cell culture supernatants and tested in human umbilical vein endothelial cell (HUVEC) proliferation and migration assays to confirm that functional protein was being made. Finally, in vivo activity was assessed by injecting 10(6) tumor cells subcutaneously into SCID mice and monitoring subsequent tumor growth. RESULTS: Purified flk-1 (0.1 micromol/L) was able to inhibit basic fibroblast growth factor (bFGF) stimulated HUVEC proliferation by 44% and VEGF-stimulated migration by 30%. In vitro growth rates for the transduced cell lines were similar to the unmodified cell line. In vivo, however, after 23 days, tumors from flk-1 expressing neuroblastoma cells were less than 33% the average volume of tumors from cells expressing only the GFP transgene (mean volume, 1.9 cm(3) v 5.8 cm(3), P<.001). GFP expression alone had no effect on tumor growth when compared with unmodified tumor cells. CONCLUSIONS: Engineered expression of flk-1, a competitive inhibitor of VEGF, by tumor cells results in the production of an inhibitor of endothelial cell proliferation and migration that greatly restricts the growth of the tumor cells in vivo. Gene therapy-mediated delivery of angiogenesis inhibitors may provide an alternative approach to treating refractory tumors such as neuroblastoma.


Asunto(s)
Terapia Genética/métodos , Neovascularización Patológica/prevención & control , Neuroblastoma/tratamiento farmacológico , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Animales , Western Blotting , Vectores Genéticos , Humanos , Ratones , Ratones SCID , Neuroblastoma/irrigación sanguínea , Neuroblastoma/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular , Transducción Genética , Células Tumorales Cultivadas/metabolismo
10.
Mol Cell Biol ; 21(2): 678-89, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11134353

RESUMEN

Jak3-deficient mice display vastly reduced numbers of lymphoid cells. Thymocytes and peripheral T cells from Jak3-deficient mice have a high apoptotic index, suggesting that Jak3 provides survival signals. Here we report that Jak3 regulates T lymphopoiesis at least in part through its selective regulation of Bax and Bcl-2. Jak3-deficient thymocytes express elevated levels of Bax and reduced levels of Bcl-2 relative to those in wild-type littermates. Notably, up-regulation of Bax in Jak3-deficient T cells is physiologically relevant, as Jak3 Bax double-null mice have marked increases in thymocyte and peripheral T-cell numbers. Rescue of T lymphopoiesis by Bax loss was selective, as mice deficient in Jak3 plus p53 or in Jak3 plus Fas remained lymphopenic. However, Bax loss failed to restore proper ratios of peripheral CD4/CD8 T cells, which are abnormally high in Jak3-null mice. Transplantation into Jak3-deficient mice of Jak3-null bone marrow transduced with a Bcl-2-expressing retrovirus also improved peripheral T-cell numbers and restored the ratio of peripheral CD4/CD8 T cells to wild-type levels. The data support the concepts that Jak kinases regulate cell survival through their selective and cell context-dependent regulation of pro- and antiapoptotic Bcl-2 family proteins and that Bax and Bcl-2 play distinct roles in T-cell development.


Asunto(s)
Regulación de la Expresión Génica , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas/genética , Linfocitos T/citología , Animales , Apoptosis , Relación CD4-CD8 , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Supervivencia Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Eliminación de Gen , Etiquetado Corte-Fin in Situ , Janus Quinasa 3 , Ratones , Ratones Noqueados , Proteínas Tirosina Quinasas/deficiencia , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Linfocitos T/metabolismo , Timo/citología , Timo/crecimiento & desarrollo , Proteína X Asociada a bcl-2
11.
Curr Gene Ther ; 1(3): 257-65, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12109141

RESUMEN

Gene transfer into stem cells has long been studied as a means by which primitive hematopoietic cells could be characterized and manipulated. While a variety of strategies have been attempted, it still remains relatively difficult to perform direct stem cell analysis. In this review, we examine recent studies using adenovirus-based vectors as a means to achieve high-level gene transfer into primitive hematopoietic cell types.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos , Células Madre Hematopoyéticas/fisiología , Transducción Genética , Animales , Técnicas de Transferencia de Gen , Humanos
12.
Med Pediatr Oncol ; 35(6): 638-40, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11107136

RESUMEN

BACKGROUND: The purpose of this study was to determine whether gene therapy-mediated delivery of an angiogenesis inhibitor, a truncated, soluble vascular endothelial growth factor receptor (Flk-1/KDR, VEGFR-2), could suppress tumor growth in a murine model of neuroblastoma. METHODS: Murine fibroblasts producing a replication-defective retrovirus encoding this mutant form of flk-1 were made. These producer cells were mixed with neuroblastoma cells and injected subcutaneously into SCID mice. Subsequent tumor growth was then measured. RESULTS: Murine neuroblastoma growth was decreased by 95% after 25 days. Similar tumor growth inhibitory effects were observed when the flk-1 producer cells were co-injected with cells from two different human neuroblastoma cell lines. CONCLUSIONS: Neuroblastoma growth can be significantly restricted in vivo with a single injection of cells that produce a retroviral vector encoding the gene for an angiogenesis inhibitor. This suggests that gene therapy-mediated delivery can be an effective alternative to chronic administration of these cytostatic anticancer agents.


Asunto(s)
Vectores Genéticos , Neovascularización Patológica , Neuroblastoma/genética , Neuroblastoma/patología , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento/genética , Receptores Mitogénicos/genética , Retroviridae , Animales , División Celular , Humanos , Ratones , Neuroblastoma/terapia , Receptores de Factores de Crecimiento Endotelial Vascular , Factores de Tiempo , Células Tumorales Cultivadas
13.
Med Pediatr Oncol ; 35(6): 712-5, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11107153

RESUMEN

BACKGROUND: Immunotherapy using cytokine-expressing tumor cells has shown promise as an anticancer strategy. We have recently begun a trial of interleukin-2 (IL-2) gene-modified allogeneic neuroblastoma cells administered in a sequence of eight injections to patients with high-risk neuroblastoma following completion of primary therapy. Six patients to date have completed treatment. PROCEDURE: We examined humoral responses to the immunizing cell line and, when available, to the patients' autologous tumor cells using an in vitro binding assay. RESULTS: Five of six patients developed a rise in antitumor antibodies to the immunizing neuroblastoma cell line following vaccination. Two of these patients had autologous tumor available; both demonstrated a humoral response to these cells as well. CONCLUSIONS: Our results demonstrate that vaccination with IL-2-expressing allogeneic tumor cells after intensive primary therapy can elicit a humoral response to the immunizing line. These antibodies are cross-reactive with the patients' own tumor cells in the two cases in which autologous cells were available. This suggests that different patients' tumors may share common antigens that can be exploited in immunotherapy strategies and supports the continued exploration of allogeneic tumor cells as tumor vaccines.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Regulación Neoplásica de la Expresión Génica , Interleucina-2/genética , Neuroblastoma/inmunología , Neuroblastoma/terapia , Adolescente , Formación de Anticuerpos , Niño , Preescolar , Humanos , Lactante , Neuroblastoma/genética , Resultado del Tratamiento , Células Tumorales Cultivadas
14.
Blood ; 96(4): 1206-14, 2000 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-10942359

RESUMEN

Limited expression of the amphotropic envelope receptor is a recognized barrier to efficient oncoretroviral vector-mediated gene transfer. Human hematopoietic cell lines and cord blood-derived CD34(+) and CD34(+), CD38(-) cell populations and the progenitors contained therein were transduced far more efficiently with oncoretroviral particles pseudotyped with the envelope protein of feline endogenous virus (RD114) than with conventional amphotropic vector particles. Similarly, human repopulating cells from umbilical cord blood capable of establishing hematopoiesis in immunodeficient mice were efficiently transduced with RD114-pseudotyped particles, whereas amphotropic particles were ineffective at introducing the proviral genome. After only a single exposure of CD34(+) cord blood cells to RD114-pseudotyped particles, all engrafted nonobese diabetic/severe combined immunodeficiency mice (15 of 15) contained genetically modified human bone marrow cells. Human cells that were positive for enhanced green fluorescent protein represented as much as 90% of the graft. The use of RD114-pseudotyped vectors may be advantageous for therapeutic gene transfer into hematopoietic stem cells. (Blood. 2000;96:1206-1214)


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Retroviridae , Proteínas del Envoltorio Viral/genética , Animales , Gatos , Diabetes Mellitus Tipo 1 , Sangre Fetal , Humanos , Ratones , Ratones Endogámicos NOD
15.
Nat Med ; 6(5): 529-35, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10802708

RESUMEN

Caspase 8 is a cysteine protease regulated in both a death-receptor-dependent and -independent manner during apoptosis. Here, we report that the gene for caspase 8 is frequently inactivated in neuroblastoma, a childhood tumor of the peripheral nervous system. The gene is silenced through DNA methylation as well as through gene deletion. Complete inactivation of CASP8 occurred almost exclusively in neuroblastomas with amplification of the oncogene MYCN. Caspase 8-null neuroblastoma cells were resistant to death receptor- and doxorubicin-mediated apoptosis, deficits that were corrected by programmed expression of the enzyme. Thus, caspase 8 acts as a tumor suppressor in neuroblastomas with amplification of MYCN.


Asunto(s)
Caspasas/genética , Amplificación de Genes , Silenciador del Gen , Genes myc , Neuroblastoma/genética , Antineoplásicos/farmacología , Apoptosis , Caspasa 8 , Caspasa 9 , Caspasas/biosíntesis , Niño , Metilación de ADN , Doxorrubicina/farmacología , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Recombinantes/biosíntesis , Retroviridae/genética , Transducción de Señal , Células Tumorales Cultivadas
16.
Gene Ther ; 7(3): 183-95, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10694794

RESUMEN

Recombinant adeno-associated viral (rAAV) vectors have been evaluated for their ability to transduce primitive hematopoietic cells. Early studies documented rAAV-mediated gene expression during progenitor derived colony formation in vitro, but studies examining genome integration and long-term gene expression in hematopoietic cells have yielded conflicting results. Such studies were performed with crude vector preparations. Using improved methodology, we have generated high titer, biologically active preparations of rAAV free of wild-type AAV (less than 1/107particles) and adenovirus. Transduction of CD34+ cells from umbilical cord blood was evaluated with a bicistronic rAAV vector encoding the green fluorescent protein (GFP) and a trimetrexate resistant variant of dihydrofolate reductase (DHFR). Freshly isolated, quiescent CD34+ cells were resistant to transduction (less than 4%), but transduction increased to 23 +/- 2% after 2 days of cytokine stimulation and was further augmented by addition of tumor necrosis factor alpha (51 +/- 4%) at a multiplicity of infection of 106. rAAV-mediated gene expression was transient in that progenitor derived colony formation was inhibited by trimetrexate. Primitive CD34+ and CD34+, CD38- subsets were sequentially transduced with a rAAV vector encoding the murine ecotropic receptor followed by transduction with an ecotropic retroviral vector encoding GFP and DHFR. Under optimal conditions 41 +/- 7% of CD34+ progenitors and 21 +/- 6% of CD34+, CD38- progenitors became trimetrexate resistant. These results document that highly purified rAAV transduce primitive human hematopoietic cells efficiently but gene expression appears to be transient. Gene Therapy (2000) 7, 183-195.


Asunto(s)
Adenoviridae/genética , Antígenos CD34/genética , Antígenos CD , Antígenos de Diferenciación/genética , Vectores Genéticos/genética , Células Madre Hematopoyéticas/fisiología , NAD+ Nucleosidasa/genética , ADP-Ribosil Ciclasa , ADP-Ribosil Ciclasa 1 , Animales , Células Cultivadas , Sangre Fetal , Técnicas de Transferencia de Gen , Virus Helper/genética , Humanos , Glicoproteínas de Membrana , Ratones , Transducción Genética/genética , Factor de Necrosis Tumoral alfa/farmacología
17.
Gene Ther ; 6(8): 1456-68, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10467370

RESUMEN

One factor limiting the ability to modify human repopulating hematopoietic cells genetically with retroviral vectors is the relatively low expression of the cognate viral receptor. We have tested sequential transduction of human hematopoietic cells with an adenoviral vector encoding the ecotropic retroviral receptor followed by transduction with an ecotropic retroviral vector. Adenoviral transduction of K562 erythroleukemia cells was highly efficiently with >95% of cells expressing the ecotropic receptor at a multiplicity of infection (MOI) of 103with a correspondingly high transduction with a retroviral vector. Ecotropic receptor expression in CD34+ cells following transduction with adenoviral vectors was increased by at least two-fold (from 20 to 48%) by replacing the RSV promoter with the CMV E1a promoter, resulting in a parallel increase in retroviral transduction efficiency. Replacing the head portion of the fiber protein in conventional adenoviral vectors (serotype 5) with the corresponding portion from an adenoviral 3 serotype resulted in ecotropic receptor expression in 60% of CD34+ cells at an MOI of 104 and a retroviral transduction of 60% of hematopoietic clonogenic progenitors. The sequential transduction strategy also resulted in efficient transduction of the primitive CD34+CD38- subset suggesting that it may hold promise for genetic modification of human hematopoietic stem cells.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/genética , Células Madre Hematopoyéticas/fisiología , Glicoproteínas de Membrana/genética , Receptores Virales/genética , Transducción Genética/genética , Animales , Antígenos CD34/metabolismo , Células Cultivadas , Humanos , Ratones , Proteínas Recombinantes de Fusión/farmacología
18.
J Pediatr Surg ; 34(5): 902-6; discussion 906-7, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10359203

RESUMEN

PURPOSE: Interleukin-12 (IL-12) is a cytokine with potent antitumor effects. The authors sought to assess its capacity to increase tumor immunogenicity when expressed by tumor cells in a murine model of neuroblastoma. METHODS: Syngeneic A/J mice were inoculated subcutaneously with 2 x 10(6) cells from a murine neuroblastoma-derived cell line (neuro-2a). In situ transduction of the neuroblastoma cells was achieved by intratumoral injection of an adenoviral vector encoding both subunits of the murine IL-12 heterodimer. Growth of the IL-12 gene-modified tumor cells was compared with untreated neuro-2a cells. Tumor immunity was assessed by rechallenging mice that had rejected their tumor with unmodified neuroblastoma cells. The contribution of cytotoxic T lymphocytes (CTLs) was evaluated through cytotoxicity assays. RESULTS: Eighteen (72%) of 25 tumor-bearing mice treated with the mlL-12 adenoviral vector exhibited tumor regression, with 12 mice (48%) completely rejecting their tumors over 2 to 3 weeks. None of the mice that had rejected their tumor and were rechallenged with unmodified neuro-2a cells subsequently developed new tumors. Pooled splenocytes from mice rejecting their tumors showed significant tumor killing (>20% cytolysis) in vitro in 51Cr release assays. CONCLUSIONS: Adenoviral-mediated IL-12 expression by tumor cells in a murine neuroblastoma model produced a significant antitumor response. Most treated tumors demonstrated at least transient regression, whereas many completely regressed. Cured mice exhibited protective immunity and CTL activity against the tumor. These data confirm the immunomodulatory efficacy of IL-12 as part of a vaccine-based antineuroblastoma strategy.


Asunto(s)
Terapia Genética , Vectores Genéticos , Inmunoterapia/métodos , Interleucina-12 , Neuroblastoma/inmunología , Neuroblastoma/terapia , Adenoviridae , Adyuvantes Inmunológicos , Animales , Citotoxicidad Inmunológica , Inmunohistoquímica , Interleucina-12/genética , Interleucina-12/farmacología , Ratones , Inducción de Remisión , Transducción Genética , Células Tumorales Cultivadas
19.
Hum Gene Ther ; 10(7): 1163-73, 1999 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-10340548

RESUMEN

Bone marrow stromal cells (MSCs) are unique mesenchymal cells that have been utilized as vehicles for the delivery of therapeutic proteins in gene therapy protocols. However, there are several unresolved issues regarding their potential therapeutic applications. These include low transduction efficiency, attenuation of transgene expression, and the technical problems associated with drug-based selection markers. To address these issues, we have developed a transduction protocol that yields high-level gene transfer into human MSCs, employing a murine stem cell virus-based bicistronic vector containing the green fluorescent protein (GFP) gene as a selectable marker. Transduction of MSCs plated at low density for 6 hr per day for 3 days with high-titer viral supernatant resulted in a gene transfer efficiency of 80+/-6% (n = 10) as measured by GFP fluorescence. Neither centrifugation nor phosphate depletion increased transduction efficiency. Assessment of amphotropic receptor (Pit-2) expression by RT-PCR demonstrated that all MSCs expressing the receptor were successfully transduced. Cell cycle distribution profiles measured by propidium iodide staining showed no correlation with the susceptibility of MSCs to transduction by the retroviral vector. Human MSCs sequentially transduced with an adenoviral vector encoding the ecotropic receptor and ecotropic retroviral vector encoding GFP demonstrated that all MSCs are susceptible to retroviral transduction. We further showed that both genes of bicistronic vector are expressed for at least 6 months in vitro and that transgene expression did not affect the growth or osteogenic differentiation potential of MSCs. Future studies will be directed toward the development of gene therapy protocols employing this strategy.


Asunto(s)
Células de la Médula Ósea , Técnicas de Transferencia de Gen , Vectores Genéticos , Proteínas Luminiscentes/genética , Retroviridae/genética , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Expresión Génica , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/metabolismo , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Células Madre , Células del Estroma , Factor de Transcripción Pit-1 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transducción Genética
20.
J Surg Res ; 83(2): 95-9, 1999 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-10329101

RESUMEN

BACKGROUND: A recent clinical trial of an antineuroblastoma vaccine used adenovirus serotype 5 (Ad5) vectors to transduce autologous tumor cells with the gene encoding IL-2. A method to improve transduction efficiency was sought to enable the use of lower viral titers, especially when in situ adenoviral-mediated tumor cell transduction is considered. MATERIALS AND METHODS: A chimeric adenoviral delivery vector was utilized in which the fiber head from adenovirus serotype 3 was incorporated into the backbone of Ad5. Since the fiber head protein is responsible for viral attachment to target cells, a different spectrum and range of infectivity might result. Both the chimeric (Av9LacZ4) and Ad5 (Av1LacZ4) vectors were constructed to carry a beta-galactosidase transgene. The relative transduction efficiency of these two vectors was then evaluated in five tumor-derived short-term neuroblastoma cultures and four established neuroblastoma cell lines. Enzyme activity was assessed using three different methods: in situ staining, flow cytometric analysis, and a quantitative assay. RESULTS: A significant improvement in transduction efficiency of the short-term neuroblastoma cultures with the new chimeric adenovector was demonstrated. A similar improvement in transduction efficiency was not observed in the established cell lines, suggesting that the cell surface receptor for the Ad 3 serotype had been lost in vitro. Increased transduction of tumor cells with N-myc amplification was also observed. CONCLUSIONS: The newly constructed chimeric adenoviral vector transduces short-term neuroblastoma cultures more efficiently than the standard Ad5 vector. This vector will permit the use of lower viral titers and may be useful in other adenoviral-based gene-therapy protocols. Increased transgene expression in N-myc-amplified cells offers possible selectivity for in situ gene delivery.


Asunto(s)
Vacunas contra el Cáncer , Interleucina-2/genética , Neuroblastoma/inmunología , Vacunas Sintéticas , Adenovirus Humanos , Amplificación de Genes , Genes myc , Vectores Genéticos , Humanos , Interleucina-2/biosíntesis , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Recombinantes de Fusión/biosíntesis , Transfección/métodos , Células Tumorales Cultivadas , beta-Galactosidasa/biosíntesis , beta-Galactosidasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...