Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cytotherapy ; 25(1): 46-58, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36396552

RESUMEN

BACKGROUND AIMS: The targeting of solid cancers with chimeric antigen receptor (CAR) T cells faces many technological hurdles, including selection of optimal target antigens. Promising pre-clinical and clinical data of CAR T-cell activity have emerged from targeting surface antigens such as GD2 and B7H3 in childhood cancer neuroblastoma. Anaplastic lymphoma kinase (ALK) is expressed in a majority of neuroblastomas at low antigen density but is largely absent from healthy tissues. METHODS: To explore an alternate target antigen for neuroblastoma CAR T-cell therapy, the authors generated and screened a single-chain variable fragment library targeting ALK extracellular domain to make a panel of new anti-ALK CAR T-cell constructs. RESULTS: A lead novel CAR T-cell construct was capable of specific cytotoxicity against neuroblastoma cells expressing low levels of ALK, but with only weak cytokine and proliferative T-cell responses. To explore strategies for amplifying ALK CAR T cells, the authors generated a co-CAR approach in which T cells received signal 1 from a first-generation ALK construct and signal 2 from anti-B7H3 or GD2 chimeric co-stimulatory receptors. The co-CAR approach successfully demonstrated the ability to avoid targeting single-antigen-positive targets as a strategy for mitigating on-target off-tumor toxicity. CONCLUSIONS: These data provide further proof of concept for ALK as a neuroblastoma CAR T-cell target.


Asunto(s)
Neuroblastoma , Receptores de Antígenos de Linfocitos T , Humanos , Receptores de Antígenos de Linfocitos T/genética , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Gangliósidos , Neuroblastoma/genética , Neuroblastoma/terapia , Linfocitos T , Inmunoterapia Adoptiva , Anticuerpos , Lógica
2.
Mol Ther Oncolytics ; 26: 429-443, 2022 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-36159778

RESUMEN

B7-H3 (CD276) has emerged as a target for cancer immunotherapy by virtue of consistent expression in many malignancies, relative absence from healthy tissues, and an emerging role as a driver of tumor immune inhibition. Recent studies have reported B7-H3 to be a suitable target for chimeric antigen receptor-modified T cell (CAR-T) therapy using CARs constructed from established anti-B7-H3 antibodies converted into single-chain Fv format (scFv). We constructed and screened binders in an scFv library to generate a new anti-B7-H3 CAR-T with favorable properties. This allowed access to numerous specificities ready formatted for CAR evaluation. Selected anti-human B7-H3 scFvs were readily cloned into CAR-T and evaluated for anti-tumor reactivity in cytotoxicity, cytokine, and proliferation assays. Two binders with divergent complementarity determining regions were found to show optimal antigen-specific cytotoxicity and cytokine secretion. One binder in second-generation CD28-CD3ζ CAR format induced sustained in vitro proliferation on repeat antigen challenge. The lead candidate CAR-T also demonstrated in vivo activity in a resistant neuroblastoma model. An empirical approach to B7-H3 CAR-T discovery through screening of novel scFv sequences in CAR-T format has led to the identification of a new construct with sustained proliferative capacity warranting further evaluation.

3.
Exp Hematol ; 57: 21-29, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28911908

RESUMEN

Integration-deficient lentiviruses (IdLVs) deliver genes effectively to tissues but are lost rapidly from dividing cells. This property can be harnessed to express transgenes transiently to manipulate cell biology. Here, we demonstrate the utility of short-term gene expression to improve functional potency of hematopoietic stem and progenitor cells (HSPCs) during transplantation by delivering HOXB4 and Angptl3 using IdLVs to enhance the engraftment of HSPCs. Constitutive overexpression of either of these genes is likely to be undesirable, but the transient nature of IdLVs reduces this risk and those associated with unsolicited gene expression in daughter cells. Transient expression led to increased multilineage hematopoietic engraftment in in vivo competitive repopulation assays without the side effects reported in constitutive overexpression models. Adult stem cell fate has not been programmed previously using IdLVs, but we demonstrate that these transient gene expression tools can produce clinically relevant alterations or be applied to investigate basic biology.


Asunto(s)
Vectores Genéticos/genética , Células Madre Hematopoyéticas/fisiología , Lentivirus/genética , Transducción Genética , Proteína 3 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina/biosíntesis , Proteínas Similares a la Angiopoyetina/genética , Animales , Linaje de la Célula , Regulación de la Expresión Génica , Genes Reporteros , Supervivencia de Injerto , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Humanos , Células K562 , Ratones , Ratones Endogámicos C57BL , Quimera por Radiación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transgenes
4.
FASEB J ; 29(9): 3876-88, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26062602

RESUMEN

Several acute monogenic diseases affect multiple body systems, causing death in childhood. The development of novel therapies for such conditions is challenging. However, improvements in gene delivery technology mean that gene therapy has the potential to treat such disorders. We evaluated the ability of the AAV9 vector to mediate systemic gene delivery after intravenous administration to perinatal mice and late-gestation nonhuman primates (NHPs). Titer-matched single-stranded (ss) and self-complementary (sc) AAV9 carrying the green fluorescent protein (GFP) reporter gene were intravenously administered to fetal and neonatal mice, with noninjected age-matched mice used as the control. Extensive GFP expression was observed in organs throughout the body, with the epithelial and muscle cells being particularly well transduced. ssAAV9 carrying the WPRE sequence mediated significantly more gene expression than its sc counterpart, which lacked the woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) sequence. To examine a realistic scale-up to larger models or potentially patients for such an approach, AAV9 was intravenously administered to late-gestation NHPs by using a clinically relevant protocol. Widespread systemic gene expression was measured throughout the body, with cellular tropisms similar to those observed in the mouse studies and no observable adverse events. This study confirms that AAV9 can safely mediate systemic gene delivery in small and large animal models and supports its potential use in clinical systemic gene therapy protocols.


Asunto(s)
Dependovirus , Feto , Vectores Genéticos , Proteínas Fluorescentes Verdes , Transducción Genética/métodos , Tropismo Viral , Animales , Femenino , Feto/citología , Feto/embriología , Feto/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Vectores Genéticos/farmacología , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Haplorrinos , Ratones , Embarazo
5.
Mol Ther ; 23(4): 737-45, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25523759

RESUMEN

Defects in perforin lead to the failure of T and NK cell cytotoxicity, hypercytokinemia, and the immune dysregulatory condition known as familial hemophagocytic lymphohistiocytosis (FHL). The only curative treatment is allogeneic hematopoietic stem cell transplantation which carries substantial risks. We used lentiviral vectors (LV) expressing the human perforin gene, under the transcriptional control of the ubiquitous phosphoglycerate kinase promoter or a lineage-specific perforin promoter, to correct the defect in different murine models. Following LV-mediated gene transfer into progenitor cells from perforin-deficient mice, we observed perforin expression in mature T and NK cells, and there was no evidence of progenitor cell toxicity when transplanted into irradiated recipients. The resulting perforin-reconstituted NK cells showed partial recovery of cytotoxicity, and we observed full recovery of cytotoxicity in polyclonal CD8(+) T cells. Furthermore, reconstituted T cells with defined antigen specificity displayed normal cytotoxic function against peptide-loaded targets. Reconstituted CD8(+) lymphoblasts had reduced interferon-γ secretion following stimulation in vitro, suggesting restoration of normal immune regulation. Finally, upon viral challenge, mice with >30% engraftment of gene-modified cells exhibited reduction of cytokine hypersecretion and cytopenias. This study demonstrates the potential of hematopoietic stem cell gene therapy as a curative treatment for perforin-deficient FHL.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Células Madre Hematopoyéticas/metabolismo , Células Asesinas Naturales/inmunología , Linfohistiocitosis Hemofagocítica/terapia , Perforina/genética , Animales , Linfohistiocitosis Hemofagocítica/inmunología , Ratones , Ratones Transgénicos , Fenotipo
6.
Blood ; 121(7): 1073-6, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23223356

RESUMEN

X-linked lymphoproliferative disease (XLP1) arises from mutations in the gene encoding SLAM-associated protein (SAP) and leads to abnormalities of NKT-cell development, NK-cell cytotoxicity, and T-dependent humoral function. Curative treatment is limited to allogeneic hematopoietic stem cell (HSC) transplantation. We tested whether HSC gene therapy could correct the multilineage defects seen in SAP(-/-) mice. SAP(-/-) murine HSCs were transduced with lentiviral vectors containing either SAP or reporter gene before transplantation into irradiated recipients. NKT-cell development was significantly higher and NK-cell cytotoxicity restored to wild-type levels in mice receiving the SAP vector in comparison to control mice. Baseline immunoglobulin levels were significantly increased and T-dependent humoral responses to NP-CGG, including germinal center formation, were restored in SAP-transduced mice.We demonstrate for the first time that HSC gene transfer corrects the cellular and humoral defects in SAP(-/-) mice providing proof of concept for gene therapy in XLP1.


Asunto(s)
Terapia Genética/métodos , Péptidos y Proteínas de Señalización Intracelular/genética , Trastornos Linfoproliferativos/inmunología , Trastornos Linfoproliferativos/terapia , Animales , Linaje de la Célula , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Trasplante de Células Madre Hematopoyéticas , Inmunidad Celular , Inmunidad Humoral , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Asesinas Naturales/inmunología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/patología , Ratones , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria
7.
Mol Ther ; 20(7): 1400-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22434141

RESUMEN

Some gene therapy strategies are compromised by the levels of gene expression required for therapeutic benefit, and also by the breadth of cell types that require correction. We designed a lentiviral vector system in which a transgene is under the transcriptional control of the short form of constitutively acting elongation factor 1α promoter (EFS) combined with essential elements of the locus control region of the ß-globin gene (ß-LCR). We show that the ß-LCR can upregulate EFS activity specifically in erythroid cells but does not alter EFS activity in myeloid or lymphoid cells. Experiments using the green fluorescent protein (GFP) reporter or the human adenosine deaminase (ADA) gene demonstrate 3-7 times upregulation in vitro but >20 times erythroid-specific upregulation in vivo, the effects of which were sustained for 1 year. The addition of the ß-LCR did not alter the mutagenic potential of the vector in in vitro mutagenesis (IM) assays although microarray analysis showed that the ß-LCR upregulates ~9% of neighboring genes. This vector design therefore combines the benefits of multilineage gene expression with high-level erythroid expression, and has considerable potential for correction of multisystem diseases including certain lysosomal storage diseases through a hematopoietic stem cell (HSC) gene therapy approach.


Asunto(s)
Células Precursoras Eritroides/metabolismo , Región de Control de Posición , Factor 1 de Elongación Peptídica/genética , Factores de Elongación de Péptidos/genética , Globinas beta/genética , Adenosina Desaminasa/genética , Animales , Línea Celular , Regulación de la Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Células Madre Hematopoyéticas , Humanos , Células Jurkat , Lentivirus/genética , Enfermedades por Almacenamiento Lisosomal/genética , Enfermedades por Almacenamiento Lisosomal/terapia , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Células U937 , Regulación hacia Arriba
8.
Toxicol Lett ; 199(3): 317-22, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20883753

RESUMEN

Hematopoietic system displays a wide spectrum of cell populations hierarchically organized in the bone marrow. Homeostasis in this system requires equilibrium between the self-renewal of the stem cells and their capacity of differentiation. Any failure on this equilibrium could lead to fatal consequences, such as the development of leukemia. Due to its rapid rate of renewal, hematopoietic tissue is a major target for antitumoral compounds and often becomes a dose limiting factor in the development of antineoplastics. Our aim was to develop an in vitro model for predicting the efficacy of antitumoral compounds on leukemic cells and their toxic effects on the healthy hematopoietic cells. The mouse myelomonocytic leukemia WEHI-3b was transduced with a lentiviral vector for expressing the green fluorescence protein. Mixed semisolid clonogenic cultures of transduced WEHI-3b and murine bone marrow cells were exposed to five pharmaceuticals: daunorubicin (positive control), atropine sulphate (negative control) and three in different stages of clinical development (trabectedin, Zalypsis(®) and PM01183). Colonies of leukemic cells were distinguishable from healthy CFU-GM under fluorescence microscope. The sensitivity of leukemic cells to daunorubicin, trabectedin, Zalypsis(®) and PM01183 was higher compared to healthy cells. The effect of a non-antitumoral compound, atropine sulphate, was the same on both populations. Our results show that this in vitro model is a valuable tool for studying the effect of antitumoral compounds in both tumoral and normal hematopoietic cells under the same toxic microenvironment and could safe time and facilitate the reduction of the number of animals used in preclinical development of pharmaceuticals.


Asunto(s)
Antineoplásicos/toxicidad , Sistema Hematopoyético/efectos de los fármacos , Leucemia/tratamiento farmacológico , Animales , Atropina/toxicidad , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Descubrimiento de Drogas , Ratones , Ratones Endogámicos BALB C
9.
PLoS One ; 5(12): e15525, 2010 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-21203397

RESUMEN

Chronic myeloid leukemia (CML) is a malignant clonal disorder of the hematopoietic system caused by the expression of the BCR/ABL fusion oncogene. Although it is well known that CML cells are genetically unstable, the mechanisms accounting for this genomic instability are still poorly understood. Because the Fanconi anemia (FA) pathway is believed to control several mechanisms of DNA repair, we investigated whether this pathway was disrupted in CML cells. Our data show that CML cells have a defective capacity to generate FANCD2 nuclear foci, either in dividing cells or after DNA damage. Similarly, human cord blood CD34(+) cells transduced with BCR/ABL retroviral vectors showed impaired FANCD2 foci formation, whereas FANCD2 monoubiquitination in these cells was unaffected. Soon after the transduction of CD34(+) cells with BCR/ABL retroviral vectors a high proportion of cells with supernumerary centrosomes was observed. Similarly, BCR/ABL induced a high proportion of chromosomal abnormalities, while mediated a cell survival advantage after exposure to DNA cross-linking agents. Significantly, both the impaired formation of FANCD2 nuclear foci, and also the predisposition of BCR/ABL cells to develop centrosomal and chromosomal aberrations were reverted by the ectopic expression of BRCA1. Taken together, our data show for the first time a disruption of the FA/BRCA pathway in BCR/ABL cells, suggesting that this defective pathway should play an important role in the genomic instability of CML by the co-occurrence of centrosomal amplification and DNA repair deficiencies.


Asunto(s)
Cromosomas/ultraestructura , Anemia de Fanconi/metabolismo , Proteínas de Fusión bcr-abl/metabolismo , Genes BRCA1 , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Antígenos CD34/biosíntesis , Línea Celular Tumoral , Centrosoma/ultraestructura , Aberraciones Cromosómicas , Reactivos de Enlaces Cruzados/farmacología , Daño del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Humanos , Microscopía Fluorescente/métodos , Retroviridae/genética
10.
Mol Ther ; 17(12): 2000-9, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19755962

RESUMEN

Human erythrocyte R-type pyruvate kinase deficiency (PKD) is a disorder caused by mutations in the PKLR gene that produces chronic nonspherocytic hemolytic anemia. Besides periodic blood transfusion and splenectomy, severe cases require bone marrow (BM) transplant, which makes this disease a good candidate for gene therapy. Here, the normal human R-type pyruvate kinase (hRPK) complementary (cDNA) was expressed in hematopoietic stem cells (HSCs) derived from pklr deficient mice, using a retroviral vector system. These mice show a similar red blood cell phenotype to that observed in human PKD. Transduced HSCs were transplanted into myeloablated adult PKD mice or in utero injected into nonconditioned PKD fetuses. In the myeloablated recipients, the hematological manifestations of PKD were completely resolved and normal percentages of late erythroid progenitors, reticulocyte and erythrocyte counts, hemoglobin levels and erythrocyte biochemistry were restored. Corrected cells preserved their rescuing capacity after secondary and tertiary transplant. When corrected cells were in utero transplanted, partial correction of the erythrocyte disease was obtained, although a very low number of corrected cells became engrafted, suggesting a different efficiency of cell therapy applied in utero. Our data suggest that transduction of human RPK cDNA in PKLR mutated HSCs could be an effective strategy in severe cases of PKD.


Asunto(s)
Anemia/prevención & control , Eritrocitos/enzimología , Terapia Genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Piruvato Quinasa/deficiencia , Piruvato Quinasa/genética , Animales , Diferenciación Celular , Células Cultivadas , Células Precursoras Eritroides/metabolismo , Femenino , Expresión Génica , Vectores Genéticos , Humanos , Isoenzimas , Masculino , Ratones , Ratones Endogámicos A , Ratones Endogámicos C57BL , Fenotipo , Transducción Genética , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...