Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
Neuroimage ; 103: 401-410, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25153699

RESUMEN

The problem of "voodoo" correlations-exceptionally high observed correlations in selected regions of the brain-is well recognized in neuroimaging. It arises when quantities of interest are estimated from the same data that was used to select them as interesting. In statistical terminology, the problem of inference following selection from the same data is that of selective inference. Motivated by the unwelcome side-effects of splitting the data- the recommended remedy-we adapt the recent developments in selective inference in order to construct confidence intervals (CIs) with good reproducibility prospects, even if selection and estimation are done with the same data. These intervals control the expected proportion of non-covered correlations in the selected voxels-the False Coverage Rate (FCR). They extend further toward zero than standard intervals, thus attenuating the impression made by highly biased observed correlations. They do so adaptively, in that they coincide with the standard CIs when far away from the selection point. We complement existing analytic proofs with a simulation, showing that the proposed intervals control the FCR in realistic social neuroscience problems. We also suggest a "confidence calibration plot", to allow the intervals to be reported in a clear and interpretable way. Applying the proposed methodology on a loss-aversion study, we demonstrate that with the sample size and selection type employed, selection bias is considerable. Finally, selective intervals are compared to the currently recommended data-splitting approach. We discover that our approach has more power and typically more informative, as no data is discarded. Computation of the intervals is implemented in an accompanying software package.


Asunto(s)
Algoritmos , Mapeo Encefálico , Encéfalo/fisiología , Sesgo , Simulación por Computador , Humanos
2.
Neuroimage ; 84: 113-21, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23988271

RESUMEN

Random effect analysis has been introduced into fMRI research in order to generalize findings from the study group to the whole population. Generalizing findings is obviously harder than detecting activation within the study group since in order to be significant, an activation has to be larger than the inter-subject variability. Indeed, detected regions are smaller when using random effect analysis versus fixed effects. The statistical assumptions behind the classic random effect model are that the effect in each location is normally distributed over subjects, and "activation" refers to a non-null mean effect. We argue that this model is unrealistic compared to the true population variability, where due to function-anatomy inconsistencies and registration anomalies, some of the subjects are active and some are not at each brain location. We propose a Gaussian-mixture-random-effect that amortizes between-subject spatial disagreement and quantifies it using the prevalence of activation at each location. We present a formal definition and an estimation procedure of this prevalence. The end result of the proposed analysis is a map of the prevalence at locations with significant activation, highlighting activation regions that are common over many brains. Prevalence estimation has several desirable properties: (a) It is more informative than the typical active/inactive paradigm. (b) In contrast to the usual display of p-values in activated regions - which trivially converge to 0 for large sample sizes - prevalence estimates converge to the true prevalence.


Asunto(s)
Artefactos , Mapeo Encefálico/métodos , Encéfalo/fisiología , Interpretación Estadística de Datos , Aumento de la Imagen/métodos , Interpretación de Imagen Asistida por Computador/métodos , Imagen por Resonancia Magnética/métodos , Algoritmos , Simulación por Computador , Humanos , Modelos Neurológicos , Modelos Estadísticos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
3.
Cancer Chemother Pharmacol ; 72(1): 241-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23719719

RESUMEN

PURPOSE: Brentuximab vedotin (ADCETRIS®), an antibody-drug conjugate, comprises an anti-CD30 antibody conjugated by a protease-cleavable linker to a microtubule-disrupting agent, monomethyl auristatin E (MMAE). In vitro studies showed that MMAE does not interfere with hERG K+ channels at clinically relevant concentrations. In pivotal phase 2 clinical trials in patients with relapsed or refractory Hodgkin lymphoma and systemic anaplastic large cell lymphoma, brentuximab vedotin has shown substantial efficacy and an acceptable safety profile. This phase 1 open-label study was designed to evaluate the effect of brentuximab vedotin on the duration of cardiac ventricular repolarization. METHODS: Patients with CD30-positive hematologic malignancies were treated with 1.8 mg/kg brentuximab vedotin by intravenous infusion every 3 weeks for up to 16 cycles. The primary endpoint was the change from baseline to Cycle 1 Days 2, 3, and 4 in the duration of ventricular repolarization using Fridericia's corrected QT interval (QTcF). RESULTS: There was no clinically meaningful change from baseline in the duration of ventricular repolarization as measured by QTcF in the 46 evaluable patients out of 52 total patients treated with brentuximab vedotin. There was no evidence of treatment-emergent cardiac safety abnormalities. Brentuximab vedotin was generally well tolerated with a response rate and an adverse event profile consistent with prior studies. CONCLUSION: There is no significant prolongation of the QT/QTc interval with brentuximab vedotin in patients with CD30-positive hematologic malignancies.


Asunto(s)
Antineoplásicos/efectos adversos , Corazón/efectos de los fármacos , Neoplasias Hematológicas/tratamiento farmacológico , Inmunoconjugados/efectos adversos , Antígeno Ki-1/metabolismo , Síndrome de QT Prolongado/inducido químicamente , Adulto , Anciano , Antineoplásicos/sangre , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Brentuximab Vedotina , Cardiotoxinas/efectos adversos , Cardiotoxinas/uso terapéutico , Hipersensibilidad a las Drogas/inmunología , Electrocardiografía Ambulatoria/efectos de los fármacos , Femenino , Corazón/fisiopatología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Neoplasias Hematológicas/sangre , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/fisiopatología , Enfermedad de Hodgkin/sangre , Enfermedad de Hodgkin/tratamiento farmacológico , Enfermedad de Hodgkin/metabolismo , Enfermedad de Hodgkin/fisiopatología , Humanos , Inmunoconjugados/sangre , Inmunoconjugados/farmacocinética , Inmunoconjugados/uso terapéutico , Antígeno Ki-1/sangre , Síndrome de QT Prolongado/fisiopatología , Masculino , Persona de Mediana Edad , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Índice de Severidad de la Enfermedad , Adulto Joven
4.
Leukemia ; 17(9): 1806-12, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12970780

RESUMEN

Patients with acute myelogenous leukemia or myelodysplastic syndrome may respond to farnesyl transferase inhibitors (FTIs) with partial or complete response rates noted in about 30% of such patients. FTIs prevent the attachment of a lipid farnesyl moiety to dependent proteins prior to their insertion into the plasma membrane and thereby prevent activity of these prenylation-dependent proteins, but their mechanism of tumor suppression remains unknown. Many patients receiving FTIs do experience myelosuppression. In this work, the in vitro effects of the FTI, R115777 on normal and leukemic hematopoiesis have been examined as have its effects on apoptosis induction and cell cycle profile in both leukemic blasts and normal CD34+ cells. R115777 was inhibitory to normal CD34+ cell proliferation and to leukemic blast cells, but did not affect long-term culture initiating cell frequency nor NOD-SCID reconstituting capacity. No induction of apoptosis or cell cycle changes were noted in AML blasts. These data suggest that myelosuppression with R115777 occurs largely at the intermediate to late progenitor stage of hematopoiesis and that cyclic use might avoid long-term marrow suppression.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Hematopoyesis/efectos de los fármacos , Leucemia/tratamiento farmacológico , Quinolonas/farmacología , Animales , Antígenos CD34/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 3 , Caspasas/metabolismo , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Farnesiltransferasa , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Poli(ADP-Ribosa) Polimerasas/metabolismo , Factores de Tiempo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/trasplante
5.
Cancer Gene Ther ; 10(4): 251-9, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12679797

RESUMEN

AIMS: Interferon-gamma (IFN-gamma) has been shown to upregulate MHC class I and II expression, and to promote generation of specific antitumor immune responses. We hypothesized that intratumoral administration of an IFN-gamma gene transfer vector facilitates its enhanced local production and may activate effector cells locally. We conducted a phase I dose-escalation study of a replication-deficient adenovirus-interferon-gamma construct (TG1041) to determine safety and tolerability of intratumoral administration, in advanced or locally recurrent melanoma. METHODS: Patients were enrolled at four successive dose levels: 10(7) infectious units (iu) (n=3), 10(8) iu (n=3), 10(9) iu (n=3), and 10(10) iu (n=2) per injection per week for 3 weeks. TG1041 was injected in the same tumor nodule weekly in each patient. Safety, toxicity, local and distant tumor responses and biologic correlates were evaluated. RESULTS: A total of 11 patients were enrolled and received the planned three injections per cycle. One patient with stable disease received a second cycle of treatment. A maximum tolerated dose was not reached in this study. No grade 4 toxicities were observed. Two grade 3 toxicities, fever and deep venous thrombosis were observed in one patient. The most frequently reported toxicities were grade 1 pain and redness at the injected site (n=8), and grade 1 fatigue (n=5) patients. Clinical changes observed at the local injected tumor site included erythema (n=5), a minor decrease in size of the injected lesion (n=5) and significant central necrosis by histopathology (n=1). Systemic effects included stable disease in one patient. Correlative studies did not reveal evidence of immunologic activity. CONCLUSION: Weekly intratumoral administration of TG1041 appears to be safe and well tolerated in patients with advanced melanoma.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Interferón gamma/genética , Melanoma/terapia , Adulto , Anciano , Femenino , Terapia Genética/efectos adversos , Vectores Genéticos/administración & dosificación , Humanos , Inyecciones Intralesiones , Interleucina-6/sangre , Interleucina-6/metabolismo , Masculino , Melanoma/diagnóstico , Melanoma/inmunología , Persona de Mediana Edad , Microglobulina beta-2/metabolismo
6.
J Virol ; 75(20): 9671-8, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11559799

RESUMEN

Cellular tRNA(Lys)(3) serves as the primer for reverse transcription of human immunodeficiency virus type 1 (HIV-1). tRNA(Lys)(3) interacts directly with HIV-1 reverse transcriptase (RT), is packaged into viral particles, and anneals to the primer-binding site (PBS) of the HIV-1 genome in order to initiate reverse transcription. Residue A58 of tRNA(Lys)(3), which lies outside the PBS-complementary region, is posttranscriptionally methylated to form 1-methyladenosine 58 (M(1)A58). This methylation is thought to serve as a pause signal for plus-strand strong-stop DNA synthesis during reverse transcription. However, formal proof that the methylation is necessary for the pausing of RT has not been obtained in vivo. In the present study, we investigated the role of tRNA(Lys)(3) residue A58 in the replication cycle of HIV-1 in living cells. We have developed a mutant tRNA(Lys)(3) derivative, tRNA(Lys)(3)A58U, in which A58 was replaced by U. This mutant tRNA was expressed in CEM cells. We demonstrate that the presence of M(1)A58 is necessary for the appropriate termination of plus-strand strong-stop DNA synthesis and that the absence of M(1)A58 allows RT to read the tRNA sequences beyond residue 58. In addition, we show that replacement of M(1)A58 with U inhibits the replication of HIV-1 in vivo. These results highlight the importance of tRNA primer residue A58 in the reverse transcription process. Inhibition of reverse transcription with mutant tRNA primers constitutes a novel approach for therapeutic intervention against HIV-1.


Asunto(s)
Adenosina/análogos & derivados , VIH-1/genética , Aminoacil-ARN de Transferencia/genética , Adenosina/metabolismo , Secuencia de Bases , Línea Celular , ADN Viral/biosíntesis , VIH-1/metabolismo , Humanos , Metilación , Mutagénesis Sitio-Dirigida , Aminoacil-ARN de Transferencia/metabolismo , Linfocitos T/virología , Transcripción Genética , Transfección , Replicación Viral
7.
Blood ; 98(2): 287-95, 2001 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-11435295

RESUMEN

Herpes simplex virus (HSV)-based vectors have favorable biologic features for gene therapy of leukemia and lymphoma. These include high transduction efficiency, ability to infect postmitotic cells, and large packaging capacity. The usefulness of HSV amplicon vectors for the transduction of primary human B-cell chronic lymphocytic leukemia (CLL) was explored. Vectors were constructed encoding beta-galactosidase (LacZ), CD80 (B7.1), or CD154 (CD40L) and were packaged using either a standard helper virus (HSVlac, HSVB7.1, and HSVCD40L) or a helper virus-free method (hf-HSVlac, hf-HSVB7.1, and hf-HSVCD40L). Both helper-containing and helper-free vector stocks were studied for their ability to transduce CLL cells, up-regulate costimulatory molecules, stimulate allogeneic T-cell proliferation in a mixed lymphocyte tumor reaction, and generate autologous cytotoxic T lymphocytes (CTLs). Although helper-containing and helper-free amplicon stocks were equivalent in their ability to transduce CLL cells, a vigorous T-cell proliferative response was obtained using cells transduced with hf-HSVB7.1 but not with HSVB7.1. CLL cells transduced with either HSVCD40L or hf-HSVCD40L were compared for their ability to up-regulate resident B7.1 and to function as T-cell stimulators. Significantly enhanced B7.1 expression in response to CD40L was observed using hf-HSVCD40L but not with HSVCD40L. CLL cells transduced with hf-HSVCD40L were also more effective at stimulating T-cell proliferation than those transduced with HSVCD40L stocks and were successful in stimulating autologous CTL activity. It is concluded that HSV amplicons are efficient vectors for gene therapy of hematologic malignancies and that helper virus-free HSV amplicon preparations are better suited for immunotherapy.


Asunto(s)
Vectores Genéticos , Herpesvirus Humano 1/genética , Inmunoterapia , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/terapia , Células Presentadoras de Antígenos , Antígeno B7-1/genética , Ligando de CD40/genética , Citocinas/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Activación de Linfocitos , Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Transfección , beta-Galactosidasa/genética
8.
Cancer Immunol Immunother ; 49(12): 649-62, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11258791

RESUMEN

In the present study we describe a novel murine tumor model in which the highly malignant murine B cell lymphoma 38C13 has been transduced with the cDNA encoding human tumor-associated antigen HER2/neu. This new cell line (38C13-HER2/neu) showed stable surfiace expression but not secretion of human HER2/neu. It also maintained expression of the idiotype (Id) of the surface immunoglobulin of 38C13, which serves as another tumor-associated antigen. Surprisingly, spontaneous tumor regression was observed following s.c. but not i.v. injection of 38C13-HER2/neu cells in immunocompetent syngeneic mice. Regression was more frequently observed with larger tumor cell challenges and was mediated through immunological mechanisms because it was not observed in syngeneic immunodeficient mice. Mice that showed complete tumor regression were immune to challenge with the parental cell line 38C13 and V1, a variant of 38C13 that does not express the Id. Immunity could be transferred with sera, suggesting that an antibody response mediated rejection and immunity. Continuously growing s.c. tumors as well as metastatic tumors obtained after the i.v. injection of 38C13-HER2/neu maintained expression of human HER2/neu, which can serve as a target for active immunotherapy. As spontaneous tumor regression has not been observed in other human murine models expressing human HER2/neu, our results illustrate the enormous differences that can exist among different murine tumors expressing the same antigen. The present model provides a useful tool for the study of the mechanisms of protective immunity to B cell lymphoma and for the evaluation of different therapeutic approaches based on the stimulation or suppression of the immune response.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Idiotipos de Inmunoglobulinas/inmunología , Linfoma de Células B/inmunología , Regresión Neoplásica Espontánea/patología , Receptor ErbB-2/metabolismo , 9,10-Dimetil-1,2-benzantraceno/farmacología , Animales , Anticuerpos Monoclonales/uso terapéutico , División Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Expresión Génica , Humanos , Inmunidad , Inmunización Pasiva , Inmunoglobulina G/inmunología , Inyecciones Intravenosas , Inyecciones Subcutáneas , Linfoma de Células B/patología , Ratones , Ratones Endogámicos C3H , Ratones Transgénicos , Trasplante de Neoplasias , Retroviridae/genética , Linfocitos T/inmunología , Transfección , Células Tumorales Cultivadas
10.
Clin Colorectal Cancer ; 1(2): 110-6, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12445369

RESUMEN

Stage II colorectal carcinoma is characterized by negative lymph node pathology as determined by conventional microscopic examination. These patients generally do not receive adjuvant therapy although 20%-30% will die from metastatic disease. To determine whether K-ras mutations at codon 12 could be used as a sensitive indicator of occult lymph node metastasis in stage II colon carcinoma, a retrospective study was performed using restriction endonuclease-mediated selective polymerase chain reaction (REMS-PCR) amplification. Of 106 colonic tumors analyzed, 46 were identified as positive for a K12-ras mutation in the primary tumor. Multiple lymph node samples from 38 of these 46 patients were examined by a sensitive nested PCR protocol for the presence of a K12-ras mutation. Of these 38 patients, 14 had 1 or more positive lymph nodes by PCR (37%) and 24 were negative for the mutation (63%). Of the 14 patients with a K12-ras mutation detected in lymph nodes, 8 died of the disease within 5 years (57%) compared to only 4 of the 24 patients with ras-negative lymph nodes (17%). The difference in time to death from disease, stratified using K12-ras status of lymph nodes, was statistically significant (P = 0.036; log-rank test). These results suggest K-ras mutation status of lymph nodes in patients with stage II colon cancer might identify a subgroup of patients who are more likely to develop recurrent and/or metastatic disease and benefit from adjuvant therapy. Larger studies are indicated to determine whether detection of K-ras mutation positivity in histologically negative lymph nodes portends a poor prognosis and to determine whether more aggressive use of adjuvant therapy is warranted.


Asunto(s)
Adenocarcinoma Mucinoso/epidemiología , Adenocarcinoma Mucinoso/genética , Adenocarcinoma/epidemiología , Adenocarcinoma/genética , Neoplasias Colorrectales/epidemiología , Neoplasias Colorrectales/genética , Genes ras/genética , Mutación/genética , Adenocarcinoma/diagnóstico , Adenocarcinoma Mucinoso/diagnóstico , Neoplasias Colorrectales/diagnóstico , Supervivencia sin Enfermedad , Humanos , Metástasis Linfática , Estadificación de Neoplasias , Reacción en Cadena de la Polimerasa , Pronóstico , Estudios Retrospectivos
12.
Dev Dyn ; 216(3): 244-56, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10590476

RESUMEN

Repair of damaged skeletal muscle fibers by muscle precursor cells (MPC) is central to the regeneration that occurs after injury or disease of muscle and is vital to the success of myoblast transplantation to treat inherited myopathies. However, we lack a detailed knowledge of the mechanisms of this muscle repair. Here, we have used a novel combination of techniques to study this process, marking MPC with nuclear-localizing LacZ and tracing their contribution to regeneration of muscle fibers after grafting into preirradiated muscle of the mdx nu/nu mouse. In this model system, there is muscle degeneration, but little or no regeneration from endogenous MPC. Incorporation of donor MPC into injected muscles was analyzed by preparing single viable muscle fibers at various times after cell implantation. Fibers were either stained immediately for beta-gal, or cultured to allow their associated satellite cells to migrate from the fiber and then stained for beta-gal. Marked myonuclei were located in discrete segments of host muscle fibers and were not incorporated preferentially at the ends of the fibers. All branches on host fibers were also found to be composed of myonuclei carrying the beta-gal marker. There was no significant movement of donor myonuclei within myofibers for up to 7 weeks after MPC implantation. Although donor-derived dystrophin was usually located coincidentally with donor myonuclei, in some fibers, the dystrophin protein had spread further along the mosaic myofibers than had the myonuclei of donor origin. In addition to repairing segments of the host fiber, the implanted MPC also gave rise to satellite cells, which may contribute to future muscle repair.


Asunto(s)
Músculo Esquelético/metabolismo , Distrofias Musculares/fisiopatología , Regeneración/fisiología , Animales , Células Cultivadas , Distrofina/metabolismo , Ratones , Ratones Desnudos , Ratones Transgénicos , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citología , Retroviridae/metabolismo , Trasplante de Tejidos
13.
Exp Cell Res ; 253(2): 523-32, 1999 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-10585276

RESUMEN

The ability to carry out gene targeting in somatic stem cells while maintaining their stem cell characteristics would have important implications for gene therapy and for the analysis of gene function. Using mouse myoblasts, we have explored this possibility by attempting to alter the promoter of a myosin heavy chain gene (MyHCIIB) characteristic of physiologically "fast" muscle so as to force its unscheduled expression in physiologically "slow" muscle fibers. Conditionally immortalized muscle precursor cells were transfected with a gene targeting construct designed to replace the MyHCIIB promoter with that for the carbonic anhydrase III gene (CAIII), which is highly expressed in slow muscle. A potentially targeted clone was isolated and differentiated in culture to form myotubes which expressed MyHCIIB. Cells from the same clone were injected into both slow and fast muscle of host mice, where they contributed to fiber formation. In slow muscle, the fibers derived from this clone did not express MyHCIIB; this may reflect an instability of the targeted MyHCIIB locus and/or a failure of the hybrid promoter to function in slow fibers in vivo. Nonetheless, we have demonstrated that a "promoter knock-in" gene targeting procedure can be used to generate unique MyHCIIB-expressing myotubes in culture and that conditionally immortalized myoblasts can be subjected to extensive passaging and genetic manipulation without losing their ability to form fibers in culture and in vivo.


Asunto(s)
Fibras Musculares de Contracción Rápida/citología , Fibras Musculares de Contracción Lenta/citología , Cadenas Pesadas de Miosina/genética , Células Madre/citología , Animales , Diferenciación Celular/genética , Línea Celular Transformada/química , Línea Celular Transformada/citología , Clonación Molecular/métodos , Expresión Génica/fisiología , Técnicas de Transferencia de Gen , Ratones , Fibras Musculares de Contracción Rápida/química , Fibras Musculares de Contracción Lenta/química , Músculo Esquelético/citología , Mutagénesis Insercional/fisiología , Plásmidos , Regiones Promotoras Genéticas/fisiología , Células Madre/química , Transfección
15.
Exp Cell Res ; 251(1): 156-65, 1999 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-10438581

RESUMEN

The vpr gene from the human immunodeficiency virus type-1 (HIV-1) encodes a 14-kDa protein that prevents cell proliferation by causing a block in the G(2) phase of the cell cycle. This cellular function of vpr is conserved in evolution because other primate lentiviruses, including HIV-2, SIV(mac), and SIV(agm) encode related genes that also induce G(2) arrest. After G(2) arrest, cells expressing vpr undergo apoptosis. The signaling pathways that result in vpr-induced cell cycle arrest and apoptosis have yet to be determined. The p53 tumor suppressor protein is involved in signaling pathways leading to cell cycle arrest and apoptosis in a variety of cell types. In this work, we examine the potential role of p53 in mediating cell cycle block and/or apoptosis by HIV-1 vpr and demonstrate that both phenomena occur independently of the presence and function of p53. Caspases are common mediators of apoptosis. We examined the potential role of caspases in mediating vpr-induced apoptosis by treating vpr-expressing cells with Boc-D-FMK, a broad spectrum, irreversible inhibitor of the caspase family. Boc-D-FMK significantly reduced the numbers of apoptotic cells induced by vpr. Therefore, we conclude that vpr-induced apoptosis is effected via the activation of caspases.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Ciclo Celular , Productos del Gen vpr/metabolismo , VIH-1/fisiología , Proteína p53 Supresora de Tumor/fisiología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Células COS , Inhibidores de Caspasas , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/efectos de la radiación , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Fragmentación del ADN , Doxorrubicina/farmacología , Activación Enzimática , Fase G2/efectos de los fármacos , Fase G2/efectos de la radiación , Rayos gamma , Productos del Gen vpr/genética , Vectores Genéticos/genética , VIH-1/genética , Humanos , Etiquetado Corte-Fin in Situ , Mecloretamina/farmacología , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana
16.
Front Biosci ; 4: D481-96, 1999 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10352135

RESUMEN

Gene therapy is a novel method under investigation for the treatment of genetic, metabolic and neurologic diseases, cancer and AIDS. The primary goal of gene therapy is to deliver a specific gene to a pre-determined target cell, and to direct expression of such a gene in a manner which will result in a therapeutic effect. Retroviral vectors have the ability to integrate in the host cell DNA irreversibly and therefore, are suitable vectors for permanent genetic modification of cells. Retrovirus-mediated gene transfer has been limited, however, by the inability of onco-retroviruses to productively infect non-dividing cells. Lentiviruses are unique among retroviruses because of their ability to infect target cells independently of their proliferation status. This chapter presents an up-to-date description of available lentiviral vectors, including vector design, applications to disease treatment and safety considerations. In addition, general aspects of the biology of lentiviruses with relevance to vector development will be discussed.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos , Lentivirus/fisiología , Genes Virales , Infecciones por VIH/terapia , VIH-1/genética , Lentivirus/genética , Lentivirus/patogenicidad , Infecciones por Lentivirus/genética , Proteínas Estructurales Virales/genética , Integración Viral
17.
Lab Anim Sci ; 49(2): 179-88, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10331548

RESUMEN

BACKGROUND AND PURPOSE: Expression of the HER2/neu proto-oncogene, a receptor-like transmembrane protein expressed at low levels on some normal cells, is markedly increased in a subset of human breast, colon, lung, and ovarian cancers. A humanized HER2/neu antibody has been tested as a therapeutic agent in several clinical trials, with promising results. We have developed a family of anti-HER2/neu fusion proteins. To evaluate the immunologic efficacy of these proteins, it is critical that tumors expressing the target antigen can grow in immunologically intact mice. METHOD: To produce murine tumors expressing human HER2/neu on the surface, CT26, MC38, and EL4 murine cell lines were transduced by use of a retroviral construct containing the cDNA encoding the human HER2/neu gene. RESULTS: Histologic features and kinetics of tumor growth in subcutaneous space of the human HER2/neu-expressing cells were similar to those of the respective parental cell lines. Intravenous inoculation with these cells induced disseminated malignant disease. Flow cytometric and immmunohistochemical analyses of freshly isolated tumors revealed in vivo expression of human HER2/neu. Secretion of antigen was not detected by use of an ELISA. CONCLUSION: Although an antibody response against the human HER2/neu antigen was observed, this response does not affect the growth rate of the HER2/neu-expressing cells. These murine models may be useful tools for evaluation of anti-cancer therapeutic approaches that target human HER2/neu.


Asunto(s)
Expresión Génica , Receptor ErbB-2/genética , Adenocarcinoma/inmunología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Citometría de Flujo , Humanos , Inmunoglobulina G/análisis , Linfoma/inmunología , Linfoma/metabolismo , Linfoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Proto-Oncogenes Mas , Receptor ErbB-2/análisis , Linfocitos T/inmunología , Transfección , Células Tumorales Cultivadas
18.
J Virol ; 73(4): 2832-40, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10074131

RESUMEN

Lentivirus vectors based on human immunodeficiency virus (HIV) type 1 (HIV-1) constitute a recent development in the field of gene therapy. A key property of HIV-1-derived vectors is their ability to infect nondividing cells. Although high-titer HIV-1-derived vectors have been produced, concerns regarding safety still exist. Safety concerns arise mainly from the possibility of recombination between transfer and packaging vectors, which may give rise to replication-competent viruses with pathogenic potential. We describe a novel lentivirus vector which is based on HIV, simian immunodeficiency virus (SIV), and vesicular stomatitis virus (VSV) and which we refer to as HIV/SIVpack/G. In this system, an HIV-1-derived genome is encapsidated by SIVmac core particles. These core particles are pseudotyped with VSV glycoprotein G. Because the nucleotide homology between HIV-1 and SIVmac is low, the likelihood of recombination between vector elements should be reduced. In addition, the packaging construct (SIVpack) for this lentivirus system was derived from SIVmac1A11, a nonvirulent SIV strain. Thus, the potential for pathogenicity with this vector system is minimal. The transduction ability of HIV/SIVpack/G was demonstrated with immortalized human lymphocytes, human primary macrophages, human bone marrow-derived CD34(+) cells, and primary mouse neurons. To our knowledge, these experiments constitute the first demonstration that the HIV-1-derived genome can be packaged by an SIVmac capsid. We demonstrate that the lentivirus vector described here recapitulates the biological properties of HIV-1-derived vectors, although with increased potential for safety in humans.


Asunto(s)
Terapia Genética , Vectores Genéticos , VIH-1/genética , Lentivirus/genética , Virus de la Inmunodeficiencia de los Simios/genética , Animales , Humanos , Ratones
19.
Blood ; 93(2): 643-54, 1999 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-9885227

RESUMEN

Herpes simplex virus amplicon vectors expressing RANTES (HSVrantes) and the T-cell costimulatory ligand B7.1 (HSVB7.1) were studied for their ability to elicit a tumor-specific T-cell response in a murine lymphoma model. HSVB7.1- and HSVrantes-transduced EL4 cells expressed high levels of B7.1 and RANTES as analyzed by flow cytometry and enzyme-linked immunosorbent assay, respectively. Inoculation of ex vivo HSVB7.1 transduced cells in syngeneic mice resulted in regression of both transduced cells and nontransduced cells inoculated contralaterally. Direct intratumoral injection of HSVB7.1 and/or HSVrantes alone or in combination into established EL4 tumors led to complete tumor regression in injected tumors as well as in nontransduced contralaterally implanted tumor, whereas control tumors or tumors injected with HSVlac expressing beta-galactosidase did not regress. Maximal protection was achieved with combined injection of HSVB7.1 and HSVrantes; mice showing tumor regression were resistant to rechallenge with parental EL4 cells, and tumor cell-specific cytolytic T-cell activity was observed in mice demonstrating regression. HSV amplicon-mediated delivery of immune effector molecules may represent a useful strategy for immunotherapy in the setting of pre-existing tumor.


Asunto(s)
Vectores Genéticos , Inmunoterapia , Linfoma de Células T/terapia , Simplexvirus/genética , Linfocitos T/inmunología , Animales , Antígeno B7-1/genética , Antígeno B7-1/inmunología , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Expresión Génica , Humanos , Linfoma de Células T/inmunología , Linfoma de Células T/patología , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Linfocitos T Citotóxicos/inmunología , Transfección , Células Tumorales Cultivadas
20.
AIDS Res Hum Retroviruses ; 14(18): 1617-24, 1998 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-9870314

RESUMEN

The natural ligands for the chemokine receptors CCR5 (RANTES, MIP-1alpha, and MIP-1beta) and CXCR4 (SDF-1) can act as potent inhibitors of infection by the human immunodeficiency virus type 1 (HIV-1) at the level of viral entry. Unlike antibody-mediated inhibition, chemokine-mediated inhibition is broadly effective. Different HIV-1 strains can utilize the same coreceptor(s) for viral entry and, therefore, can be blocked by the same chemokine(s). HIV-1 strains that are highly resistant to neutralization by V3-specific antibodies are sensitive to inhibition by chemokines. Therefore, the use of chemokine-derived molecules constitutes a potential therapeutic approach to prevent infection by HIV-1. We have generated a fusion protein between RANTES and human IgG3 (RANTES-IgG3). The effectiveness of RANTES-IgG3 inhibition of infection by HIV-1 was similar to that of rRANTES. Inhibition of HIV-1 by RANTES-IgG3 was specific for CCR5-dependent but not CXCR4-dependent HIV-1 isolates. Fusion of a chemokine to an IgG moiety offers two desirable properties with respect to the recombinant chemokine alone. First, IgG fusion proteins have extended half-lives in vivo. Second, molecules with IgG heavy chain moieties may be able to cross the placenta and potentially induce fetal protection.


Asunto(s)
Quimiocina CCL5/inmunología , VIH-1/inmunología , Inmunoglobulina G/inmunología , Proteínas Recombinantes de Fusión/inmunología , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , VIH-1/fisiología , Humanos , Fusión de Membrana/inmunología , Pruebas de Neutralización , Receptores CXCR4/metabolismo , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...