Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Blood ; 127(1): 122-31, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26531164

RESUMEN

T-cell-directed killing of tumor cells using bispecific antibodies is a promising approach for the treatment of hematologic malignancies. Here we describe our preclinical work with a dual-affinity retargeting (DART) molecule generated from antibodies to CD3 and CD123, designed to redirect T cells against acute myeloid leukemia blasts. The CD3×CD123 DART (also referred to as MGD006/S80880) consists of 2 independent polypeptides, each composed of the VH of 1 antibody in tandem with the VL of the other antibody. The target antigen CD123 (interleukin 3RA) is highly and differentially expressed in acute myeloid leukemia (AML) blasts compared with normal hematopoietic stem and progenitor cells. In this study we demonstrate that the CD3×CD123 DART binds to both human CD3 and CD123 to mediate target-effector cell association, T-cell activation, proliferation, and receptor diversification. The CD3×CD123 DART also induces a dose-dependent killing of AML cell lines and primary AML blasts in vitro and in vivo. These results provide the basis for testing the CD3×CD123 DART in the treatment of patients with CD123(+) AML.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Apoptosis , Complejo CD3/inmunología , Subunidad alfa del Receptor de Interleucina-3/inmunología , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/terapia , Linfocitos T/inmunología , Animales , Complejo CD3/metabolismo , Proliferación Celular , Citometría de Flujo , Genes Codificadores de la Cadena alfa de los Receptores de Linfocito T/genética , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Técnicas para Inmunoenzimas , Subunidad alfa del Receptor de Interleucina-3/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Mol Cancer ; 13: 50, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24602489

RESUMEN

BACKGROUND: Drug resistance is a significant problem in the treatment of ovarian cancer and can be caused by multiple mechanisms. Inhibition of apoptosis by the inhibitor of apoptosis proteins (IAPs) represents one such mechanism, and can be overcome by a mitochondrial protein called second mitochondria-derived activator of caspases (SMAC). We have previously shown that the ligands of sigma-2 receptors effectively induce tumor cell death. Additionally, because sigma-2 receptors are preferentially expressed in tumor cells, their ligands provide an effective mechanism for selective anti-cancer therapy. METHODS: In the current work, we have improved upon the previously described sigma-2 ligand SW43 by conjugating it to a pro-apoptotic small molecule SMAC mimetic SW IV-52, thus generating the novel cancer therapeutic SW IV-134. The new cancer drug was tested for receptor selectivity and tumor cell killing activity in vitro and in vivo. RESULTS: We have shown that SW IV-134 retained adequate sigma-2 receptor binding affinity in the context of the conjugate and potently induced cell death in ovarian cancer cells. The cell death induced by SW IV-134 was significantly greater than that observed with either SW43 or SW IV-52 alone and in combination. Furthermore, the intraperitoneal administration of SW IV-134 significantly reduced tumor burden and improved overall survival in a mouse xenograft model of ovarian cancer without causing significant adverse effects to normal tissues. Mechanistically, SW IV-134 induced degradation of cIAP-1 and cIAP-2 leading to NF-қB activation and TNFα-dependent cell death. CONCLUSIONS: Our findings suggest that coupling sigma-2 ligands to SMAC peptidomimetics enhances their effectiveness while maintaining the cancer selectivity. This encouraging proof-of-principle preclinical study supports further development of tumor-targeted small peptide mimetics via ligands to the sigma-2 receptor for future clinical applications.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Compuestos de Azabiciclo/química , Compuestos de Azabiciclo/farmacología , Oligopéptidos/química , Oligopéptidos/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Receptores sigma/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Western Blotting , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Femenino , Xenoinjertos , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Ligandos , Ratones , Ratones SCID , Proteínas Mitocondriales/química , Terapia Molecular Dirigida/métodos , Neoplasias Ováricas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Blood ; 120(19): 4093-103, 2012 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-22972985

RESUMEN

The clinical goal of allogeneic hematopoietic stem cell transplantation (allo-HSCT) is to minimize GVHD while maintaining GvL. Here, we show that interferon γ receptor-deficient (IFNγR(-/-)) allogeneic Tconv, which possess normal alloreactivity and cytotoxicity, induce significantly less GVHD than wild-type (WT) Tconv. This effect is mediated by altered trafficking of IFNγR(-/-) Tconv to GVHD target organs, especially the gastrointestinal (GI) tract. We show that the chemokine receptor CXCR3 is induced via IFNγR-mediated signaling and partially contributes to the trafficking of WT Tconv to GVHD target organs. Indeed, CXCR3(-/-) Tconv recapitulate the reduced GVHD potential of IFNγR(-/-) Tconv in a minor-mismatched GVHD model. Most importantly, IFNγR(-/-) (and CXCR3(-/-)) Tconv mediate a robust and beneficial GvL effect. In addition, we show that IFNγR(-/-) regulatory T cells (Tregs) are fully suppressive in vitro although defective in suppressor function in vivo and that WT Tregs suppress GVHD in vivo only when allogeneic Tconv produce interferon γ (IFNγ), suggesting that the IFNγR signaling pathway is the major mechanism for both Tregs and Tconv to migrate to GVHD target organs. Finally, pharmacologic inhibition of IFNγR signaling with inhibitors of JAK1/JAK2, which are mediators of IFNγR signaling, results in the decreased expression of CXCR3 and reduced GVHD and improved survival after allo-HSCT and this effect is mediated by altered trafficking of Tconv to GVHD target organs.


Asunto(s)
Movimiento Celular/inmunología , Enfermedad Injerto contra Huésped/inmunología , Receptores de Interferón/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Células Cultivadas , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/metabolismo , Enfermedad Injerto contra Huésped/mortalidad , Trasplante de Células Madre Hematopoyéticas , Humanos , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Ratones , Ratones Transgénicos , Nitrilos , Pirazoles/farmacología , Pirimidinas , Receptores CXCR3/genética , Receptores CXCR3/inmunología , Receptores CXCR3/metabolismo , Receptores de Interferón/genética , Transducción de Señal/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Trasplante Homólogo/efectos adversos , Trasplante Homólogo/inmunología , Receptor de Interferón gamma
4.
BMC Cancer ; 14: 35, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24447304

RESUMEN

BACKGROUND: The targeted delivery of cancer therapeutics represents an ongoing challenge in the field of drug development. TRAIL is a promising cancer drug but its activity profile could benefit from a cancer-selective delivery mechanism, which would reduce potential side effects and increase treatment efficiencies. We recently developed the novel TRAIL-based drug platform TR3, a genetically fused trimer with the capacity for further molecular modifications such as the addition of tumor-directed targeting moieties. MUC16 (CA125) is a well characterized biomarker in several human malignancies including ovarian, pancreatic and breast cancer. Mesothelin is known to interact with MUC16 with high affinity. In order to deliver TR3 selectively to MUC16-expressing cancers, we investigated the possibility of targeted TR3 delivery employing the high affinity mesothelin/MUC16 ligand/receptor interaction. METHODS: Using genetic engineering, we designed the novel cancer drug Meso-TR3, a fusion protein between native mesothelin and TR3. The recombinant proteins were produced with mammalian HEK293T cells. Meso-TR3 was characterized for binding selectivity and killing efficacy against MUC16-positive cancer cells and controls that lack MUC16 expression. Drug efficacy experiments were performed in vitro and in vivo employing an intraperitoneal xenograft mouse model of ovarian cancer. RESULTS: Similar to soluble mesothelin itself, the strong MUC16 binding property was retained in the Meso-TR3 fusion protein. The high affinity ligand/receptor interaction was associated with a selective accumulation of the cancer drug on MUC16-expressing cancer targets and directly correlated with increased killing activity in vitro and in a xenograft mouse model of ovarian cancer. The relevance of the mesothelin/MUC16 interaction for attaching Meso-TR3 to the cancer cells was verified by competitive blocking experiments using soluble mesothelin. Mechanistic studies using soluble DR5-Fc and caspase blocking assays confirmed engagement of the extrinsic death receptor pathway. Compared to non-targeted TR3, Meso-TR3 displayed a much reduced killing potency on cells that lack MUC16. CONCLUSIONS: Soluble Meso-TR3 targets the cancer biomarker MUC16 in vitro and in vivo. Following attachment to the tumor via surface bound MUC16, Meso-TR3 acquires full activation with superior killing profiles compared to non-targeted TR3, while its bioactivity is substantially reduced on cells that lack the tumor marker. This prodrug phenomenon represents a highly desirable property because it has the potential to enhance cancer killing with fewer side-effects than non-targeted TRAIL-based therapeutics. Thus, further exploration of this novel fusion protein is warranted as a possible therapeutic for patients with MUC16-positive malignancies.


Asunto(s)
Antineoplásicos/farmacología , Antígeno Ca-125/metabolismo , Portadores de Fármacos , Proteínas Ligadas a GPI/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Animales , Antineoplásicos/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Células HEK293 , Células HeLa , Humanos , Células Jurkat , Ligandos , Mesotelina , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Unión Proteica , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Curr Biol ; 16(22): 2222-7, 2006 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-17113386

RESUMEN

In eukaryotes, distinct regions of the genome are packaged as euchromatin (less condensed, more active) or heterochromatin (condensed, silenced). Studies in yeast, plants, and flies suggest that RNA interference (RNAi) is linked to heterochromatin formation and transcriptional silencing of transposable element (TE) sequences. We previously reported that insertion of a mobile hsp70-white reporter within 10 kb of a 1360 element on chromosome four of Drosophila melanogaster correlates with variegation (silencing). Here, we report small RNAs (approximately 23 nt) corresponding to 1360, indicating processing by the RNAi machinery. To directly test the ability of 1360 to silence a nearby gene in vivo, we introduced a P element construct carrying a single copy of 1360 upstream of the hsp70-white reporter into flies. This 1360 element contributes to HP1-dependent variegation at a pericentric insertion site, as demonstrated by a decrease in silencing after FLP-mediated removal of 1360. In euchromatin, 1360 is not sufficient to induce silencing, suggesting that proximity to pericentric heterochromatin and/or a high local TE density contributes to heterochromatin formation. Silencing of the 1360, hsp70-white reporter is sensitive to mutations in RNAi components. Our results implicate 1360 as a target for sequence-specific heterochromatic silencing through an RNAi-dependent mechanism.


Asunto(s)
Elementos Transponibles de ADN/genética , Drosophila melanogaster/genética , Silenciador del Gen , Heterocromatina/genética , Interferencia de ARN , Animales , Northern Blotting , Cartilla de ADN , Proteínas de Drosophila , Genes Reporteros/genética , Proteínas HSP70 de Choque Térmico/genética , Proteínas Nucleares , Proteínas de Unión al ARN
6.
Mol Imaging ; 8(5): 245-53, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19796602

RESUMEN

Small-animal tumor models are essential for developing translational therapeutic strategies in oncology research, with imaging having an increasingly important role. Magnetic resonance imaging (MRI) offers tumor localization, volumetric measurement, and the potential for advanced physiologic imaging but is less well suited to high-throughput studies and has limited capacity to assess early tumor growth. Bioluminescence imaging (BLI) identifies tumors early, monitors tumor growth, and efficiently measures response to therapeutic intervention. Generally, BLI signals have been found to correlate well with magnetic resonance measurements of tumor volume. However, in our studies of small-animal models of malignant brain tumors, we have observed specific instances in which BLI data do not correlate with corresponding MRIs. These observations led us to hypothesize that use of BLI and MRI together, rather than in isolation, would allow more effective and efficient measures of tumor growth in preclinical studies. Herein we describe combining BLI and MRI studies to characterize tumor growth in a mouse model of glioblastoma. The results led us to suggest a cost-effective, multimodality strategy for selecting cohorts of animals with similar tumor growth patterns that improves the accuracy of longitudinal in vivo measurements of tumor growth and treatment response in preclinical therapeutic studies.


Asunto(s)
Neoplasias Encefálicas/patología , Diagnóstico por Imagen/métodos , Mediciones Luminiscentes/métodos , Imagen por Resonancia Magnética/métodos , Animales , Línea Celular Tumoral , Femenino , Glioblastoma/patología , Ratones , Ratones Endogámicos BALB C
7.
Community Pract ; 82(4): 20-3, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19397079

RESUMEN

Health visitors identified fathers as a marginalised, difficult-to-reach group when aiming for universal implementation of a public health programme to prevent non-accidental head injury (NAHI) in babies. Follow-on research with 30 fathers from disadvantaged backgrounds, including some in prison, explored barriers and facilitating factors to preventing NAHI through focus groups and interviews. Fathers expressed both responsibility and helplessness in managing a baby, but many felt excluded from gaining skills and knowledge by healthcare staff. Barriers to implementing a prevention programme included a lack of knowledge about head injuries in babies and poor understanding of prevention as a reduction of risk factors. Fear of blame for a head injury could lead to injury concealment, indicating a taboo subject. Facilitators for fathers to learn about preventing head injuries in babies included concerns of their masculinity being at odds with the frailty of the newborn, recognition of fathers' needs to be valued in their own right, and a need for individualised plans for gaining information and increasing confidence. The modifiable barriers to prevention, from the fathers' perspectives, add to the theoretical and applied evidence base for the prevention of NAHI.


Asunto(s)
Padre , Educación en Salud , Evaluación de Necesidades , Carencia Psicosocial , Síndrome del Bebé Sacudido/prevención & control , Enfermería en Salud Comunitaria , Conocimientos, Actitudes y Práctica en Salud , Humanos , Lactante , Masculino , Prisioneros , Gales
8.
Mol Cell Biol ; 24(18): 8210-20, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15340080

RESUMEN

The heterochromatic domains of Drosophila melanogaster (pericentric heterochromatin, telomeres, and the fourth chromosome) are characterized by histone hypoacetylation, high levels of histone H3 methylated on lysine 9 (H3-mK9), and association with heterochromatin protein 1 (HP1). While the specific interaction of HP1 with both H3-mK9 and histone methyltransferases suggests a mechanism for the maintenance of heterochromatin, it leaves open the question of how heterochromatin formation is targeted to specific domains. Expression characteristics of reporter transgenes inserted at different sites in the fourth chromosome define a minimum of three euchromatic and three heterochromatic domains, interspersed. Here we searched for cis-acting DNA sequence determinants that specify heterochromatic domains. Genetic screens for a switch in phenotype demonstrate that local deletions or duplications of 5 to 80 kb of DNA flanking a transposon reporter can lead to the loss or acquisition of variegation, pointing to short-range cis-acting determinants for silencing. This silencing is dependent on HP1. A switch in transgene expression correlates with a switch in chromatin structure, judged by nuclease accessibility. Mapping data implicate the 1360 transposon as a target for heterochromatin formation. We propose that heterochromatin formation is initiated at dispersed repetitive elements along the fourth chromosome and spreads for approximately 10 kb or until encountering competition from a euchromatic determinant.


Asunto(s)
Drosophila melanogaster/genética , Heterocromatina/genética , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Mapeo Cromosómico , Cromosomas/genética , ADN/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Anomalías del Ojo/genética , Femenino , Silenciador del Gen , Genes de Insecto , Genes Reporteros , Prueba de Complementación Genética , Heterocromatina/metabolismo , Masculino , Mutación , Fenotipo , Eliminación de Secuencia , Secuencias Repetidas en Tándem
9.
Am J Obstet Gynecol ; 194(4): 1137-42; discussion 1142-4, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16580313

RESUMEN

OBJECTIVE: The purpose of this study was to test the hypothesis that placental samples that are obtained by needle aspiration ex vivo are useful for the determination of villus gene expression. STUDY DESIGN: Placental biopsy was performed with a spinal needle after uncomplicated deliveries. Villi were inspected microscopically, and RNA was extracted and analyzed with capillary electrophoresis. Gene expression was determined with quantitative polymerase chain reaction. RESULTS: We obtained more placental villous fragments per aspiration using a 20-gauge needle (5.2 +/- 1.8 fragments) than with a 22-gauge needle (3.3 +/- 1.6 fragments; P < .01). RNA quality was adequate, based on the 28S and 18S recombinant RNA bands, with a mean 260/280 ratio of 1.88. The amount of extracted RNA correlated with the number of villous fragments per aspirate. Importantly, the expression of NDRG1 and hPL, both markedly altered in hypoxia, was consistent between villi that were obtained by either needle or standard biopsy. CONCLUSION: Placental samples that are obtained by ex vivo needle aspiration are useful for the extraction of RNA and for the determination of villous gene expression.


Asunto(s)
Biopsia con Aguja , Placenta/química , ARN/análisis , Proteínas de Ciclo Celular , Femenino , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lactógeno Placentario/genética , Reacción en Cadena de la Polimerasa , Proteínas/genética , ARN/aislamiento & purificación
10.
Clin Cancer Res ; 22(3): 596-608, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26423796

RESUMEN

PURPOSE: Anti-CD20 monoclonal antibodies (mAb) are an important immunotherapy for B-cell lymphoma, and provide evidence that the immune system may be harnessed as an effective lymphoma treatment approach. ALT-803 is a superagonist IL-15 mutant and IL-15Rα-Fc fusion complex that activates the IL-15 receptor constitutively expressed on natural killer (NK) cells. We hypothesized that ALT-803 would enhance anti-CD20 mAb-directed NK-cell responses and antibody-dependent cellular cytotoxicity (ADCC). EXPERIMENTAL DESIGN: We tested this hypothesis by adding ALT-803 immunostimulation to anti-CD20 mAb triggering of NK cells in vitro and in vivo. Cell lines and primary human lymphoma cells were utilized as targets for primary human NK cells. Two complementary in vivo mouse models were used, which included human NK-cell xenografts in NOD/SCID-γc (-/-) mice. RESULTS: We demonstrate that short-term ALT-803 stimulation significantly increased degranulation, IFNγ production, and ADCC by human NK cells against B-cell lymphoma cell lines or primary follicular lymphoma cells. ALT-803 augmented cytotoxicity and the expression of granzyme B and perforin, providing one potential mechanism for this enhanced functionality. Moreover, in two distinct in vivo B-cell lymphoma models, the addition of ALT-803 to anti-CD20 mAb therapy resulted in significantly reduced tumor cell burden and increased survival. Long-term ALT-803 stimulation of human NK cells induced proliferation and NK-cell subset changes with preserved ADCC. CONCLUSIONS: ALT-803 represents a novel immunostimulatory drug that enhances NK-cell antilymphoma responses in vitro and in vivo, thereby supporting the clinical investigation of ALT-803 plus anti-CD20 mAbs in patients with indolent B-cell lymphoma.


Asunto(s)
Antineoplásicos/farmacología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Proteínas/farmacología , Receptores de IgG/metabolismo , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Línea Celular Tumoral , Citotoxicidad Inmunológica/efectos de los fármacos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Granzimas/genética , Granzimas/metabolismo , Humanos , Interferón gamma/biosíntesis , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/patología , Ratones , Ratones Noqueados , Perforina/genética , Perforina/metabolismo , Proteínas Recombinantes de Fusión , Rituximab/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA