Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Int J Cancer ; 134(10): 2383-92, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24154990

RESUMEN

Radiation therapy (RT) continues to be a cornerstone in the treatment for many cancers. Unfortunately, not all individuals respond effectively to RT resulting clinically in two groups consisting of nonresponders (progressive disease) and responders (tumor control/cure). The mechanisms that govern the outcome of radiotherapy are poorly understood. Interestingly, a new paradigm has emerged demonstrating that the immune system mediates many of the antitumor effects of RT. Therefore, we hypothesized that the immune response following RT may dictate the efficacy of treatment. To examine this, we developed a tumor model that mirrors this clinically relevant phenomenon in which mice bearing Colon38, a colon adenocarcinoma, were treated locally with 15Gy RT resulting in both nonresponders and responders. More importantly, we were able to distinguish responders from nonresponders as early as 4 days post-RT allowing for the unique opportunity to identify critical events that ultimately determined the effectiveness of therapy. Intratumoral immune cells and interferon-gamma were increased in responsive tumors and licensed CD8 T cells to exhibit lytic activity against tumor cells, a response that was diminished in tumors refractory to RT. Combinatorial treatment with RT and the immunomodulatory cytokine IL-12 resulted in complete remission of cancer in 100% of cases compared to a cure rate of only 12% with RT alone. Similar data were obtained when IL-12 was delivered by microspheres. Therefore, the efficacy of RT may depend on the strength of the immune response induced after radiotherapy. Additionally, immunotherapy that further stimulates the immune cells may enhance the effectiveness of RT.


Asunto(s)
Adenocarcinoma/radioterapia , Linfocitos T CD8-positivos/efectos de la radiación , Neoplasias del Colon/radioterapia , Citotoxicidad Inmunológica/efectos de la radiación , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/inmunología , Análisis de Varianza , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Quimioradioterapia , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/inmunología , Citotoxicidad Inmunológica/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/patología , Sistema Inmunológico/efectos de la radiación , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-12/farmacología , Ratones , Resultado del Tratamiento
2.
Cancer Immunol Immunother ; 63(3): 259-71, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24357146

RESUMEN

The need for an intact immune system for cancer radiation therapy to be effective suggests that radiation not only acts directly on the tumor but also indirectly, through the activation of host immune components. Recent studies demonstrated that endogenous type I interferons (type I IFNs) play a role in radiation-mediated anti-tumor immunity by enhancing the ability of dendritic cells to cross-prime CD8(+) T cells. However, it is still unclear to what extent endogenous type I IFNs contribute to the recruitment and function of CD8(+) T cells. Little is also known about the effects of type I IFNs on myeloid cells. In the current study, we demonstrate that type I and type II IFNs (IFN-γ) are both required for the increased production of CXCL10 (IP-10) chemokine by myeloid cells within the tumor after radiation treatment. Radiation-induced intratumoral IP-10 levels in turn correlate with tumor-infiltrating CD8(+) T cell numbers. Moreover, type I IFNs promote potent tumor-reactive CD8(+) T cells by directly affecting the phenotype, effector molecule production, and enhancing cytolytic activity. Using a unique inducible expression system to increase local levels of IFN-α exogenously, we show here that the capacity of radiation therapy to result in tumor control can be enhanced. Our preclinical approach to study the effects of local increase in IFN-α levels can be used to further optimize the combination therapy strategy in terms of dosing and scheduling, which may lead to better clinical outcome.


Asunto(s)
Linfocitos T CD8-positivos/efectos de la radiación , Interferón-alfa/metabolismo , Neoplasias Mamarias Animales/radioterapia , Melanoma Experimental/inmunología , Melanoma Experimental/radioterapia , Células Mieloides/efectos de los fármacos , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular/efectos de la radiación , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Citotoxicidad Inmunológica/genética , Citotoxicidad Inmunológica/efectos de la radiación , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Interferón-alfa/genética , Interferón-alfa/farmacología , Interferón gamma/genética , Interferón gamma/metabolismo , Recuento de Linfocitos , Neoplasias Mamarias Animales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Trasplante de Neoplasias
3.
Immunology ; 138(3): 280-92, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23198850

RESUMEN

The tumour microenvironment is complex containing not only neoplastic cells but also a variety of host cells. The heterogeneous infiltrating immune cells include subsets of cells with opposing functions, whose activities are mediated either directly or through the cytokines they produce. Systemic delivery of cytokines such as interleukin-2 ( IL-2) has been used clinically to enhance anti-tumour responses, but these molecules are generally thought to have evolved to act locally in a paracrine fashion. In this study we examined the effect of local production of IL-2 on the growth and the immune response to B16 melanoma cells. We found that the local production of IL-2 enhances the number of interferon-γ-expressing CD8 T and natural killer cells in the tumour, as well as inducing expression of vascular cell adhesion molecule 1 on tumour vessels. These responses were largely absent in interferon-γ knockout mice. The expression of IL-2 in the tumour microenvironment decreases tumour growth despite also enhancing Foxp3(+)  CD4(+) regulatory T cells and anti-inflammatory cytokines such as IL-10. Higher levels of IL-2 in the tumour microenvironment eliminated the progressive growth of the B16 cells in vivo, and this inhibition was dependent on the presence of either T cells or, to a lesser extent, natural killer cells. Surprisingly however, the B16 tumours were not completely eliminated but instead were controlled for an extended period of time, suggesting that a form of tumour dormancy was established.


Asunto(s)
Interleucina-2/metabolismo , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Expresión Génica , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-2/genética , Células Asesinas Naturales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/patología , Ratones , Ratones Desnudos , Carga Tumoral/genética , Carga Tumoral/inmunología , Microambiente Tumoral/genética , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
4.
Proc Natl Acad Sci U S A ; 106(48): 20435-9, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19918064

RESUMEN

Dengue virus (DENV), a mosquito-borne flavivirus, is a major public health threat. The virus poses risk to 2.5 billion people worldwide and causes 50 to 100 million human infections each year. Neither a vaccine nor an antiviral therapy is currently available for prevention and treatment of DENV infection. Here, we report a previously undescribed adenosine analog, NITD008, that potently inhibits DENV both in vitro and in vivo. In addition to the 4 serotypes of DENV, NITD008 inhibits other flaviviruses, including West Nile virus, yellow fever virus, and Powassan virus. The compound also suppresses hepatitis C virus, but it does not inhibit nonflaviviruses, such as Western equine encephalitis virus and vesicular stomatitis virus. A triphosphate form of NITD008 directly inhibits the RNA-dependent RNA polymerase activity of DENV, indicating that the compound functions as a chain terminator during viral RNA synthesis. NITD008 has good in vivo pharmacokinetic properties and is biologically available through oral administration. Treatment of DENV-infected mice with NITD008 suppressed peak viremia, reduced cytokine elevation, and completely prevented the infected mice from death. No observed adverse effect level (NOAEL) was achieved when rats were orally dosed with NITD008 at 50 mg/kg daily for 1 week. However, NOAEL could not be accomplished when rats and dogs were dosed daily for 2 weeks. Nevertheless, our results have proved the concept that a nucleoside inhibitor could be developed for potential treatment of flavivirus infections.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/metabolismo , Dengue/tratamiento farmacológico , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Viremia/tratamiento farmacológico , Adenosina/química , Animales , Antivirales/farmacocinética , Antivirales/uso terapéutico , Chlorocebus aethiops , Perros , Ensayo de Inmunoadsorción Enzimática , Femenino , Masculino , Ratones , Estructura Molecular , Nivel sin Efectos Adversos Observados , Ratas , Células Vero
5.
J Virol ; 84(11): 5678-86, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20237086

RESUMEN

Dengue virus (DENV) is the most prevalent mosquito-borne viral pathogen in humans. Neither vaccine nor antiviral therapy is currently available for DENV. We report here that N-sulfonylanthranilic acid derivatives are allosteric inhibitors of DENV RNA-dependent RNA polymerase (RdRp). The inhibitor was identified through high-throughput screening of one million compounds using a primer extension-based RdRp assay [substrate poly(C)/oligo(G)(20)]. Chemical modification of the initial "hit" improved the compound potency to an IC(50) (that is, a concentration that inhibits 50% RdRp activity) of 0.7 microM. In addition to suppressing the primer extension-based RNA elongation, the compound also inhibited de novo RNA synthesis using a DENV subgenomic RNA, but at a lower potency (IC(50) of 5 microM). Remarkably, the observed anti-polymerase activity is specific to DENV RdRp; the compound did not inhibit WNV RdRp and exhibited IC(50)s of >100 microM against hepatitis C virus RdRp and human DNA polymerase alpha and beta. UV cross-linking and mass spectrometric analysis showed that a photoreactive inhibitor could be cross-linked to Met343 within the RdRp domain of DENV NS5. On the crystal structure of DENV RdRp, Met343 is located at the entrance of RNA template tunnel. Biochemical experiments showed that the order of addition of RNA template and inhibitor during the assembly of RdRp reaction affected compound potency. Collectively, the results indicate that the compound inhibits RdRp through blocking the RNA tunnel. This study has provided direct evidence to support the hypothesis that allosteric pockets from flavivirus RdRp could be targeted for antiviral development.


Asunto(s)
Antivirales/química , Dengue/tratamiento farmacológico , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ortoaminobenzoatos/farmacología , Sitio Alostérico , Animales , Antivirales/farmacología , Sitios de Unión , Virus del Dengue/enzimología , Evaluación Preclínica de Medicamentos , Concentración 50 Inhibidora , ARN Viral/antagonistas & inhibidores , Ácidos Sulfínicos , ortoaminobenzoatos/uso terapéutico
6.
Virology ; 410(1): 240-7, 2011 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-21131015

RESUMEN

It remains unclear whether antibody-dependent-enhancement (ADE) of dengue infection merely augments viral attachment and entry through Fcγ receptors or immune complex binding to Fcγ receptors triggers an intrinsic signaling cascade that changes the viral permissiveness of the cell. Using human dengue-immune sera and novel human monoclonal antibodies against dengue in combination with virologic and immunologic techniques, we found that ADE infection increased the proportion of infected primary human monocytes modestly from 0.2% ± 0.1% (no Ab) to 1.7% ± 1.6% (with Ab) but the total virus output markedly from 2 ± 2 (× 10(3)) FFU to 120 ± 153 (× 10(3))FFU. However, this increased virus production was not associated with a reduced secretion of type I interferon or an elevated secretion of anti-inflammatory cytokine, IL-10. These results demonstrate that the regulation of virus production in ADE infection of primary human monocytes is more complex than previously appreciated.


Asunto(s)
Anticuerpos Antivirales/fisiología , Virus del Dengue/fisiología , Interferón Tipo I/metabolismo , Monocitos/fisiología , Monocitos/virología , Animales , Células Cultivadas , Chlorocebus aethiops , Virus del Dengue/inmunología , Regulación de la Expresión Génica/fisiología , Humanos , Interferón Tipo I/genética , Interleucinas/genética , Interleucinas/metabolismo , Células Vero , Acoplamiento Viral , Internalización del Virus
7.
Virology ; 389(1-2): 8-19, 2009 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-19419745

RESUMEN

We performed a focused siRNA screen in an A549 dengue type 2 New Guinea C subgenomic replicon cell line (Rluc-replicon) that contains a Renilla luciferase cassette. We found that siRNA mediated knock down of mevalonate diphospho decarboxylase (MVD) inhibited viral replication of the Rluc-replicon and DEN-2 NGC live virus replication in A549 cells. When the Rluc-replicon A459 cells were grown in delipidated media the replicon expression was suppressed and MVD knock down could further sensitize Renilla expression. Hymeglusin and zaragozic acid A could inhibit DEN-2 NGC live virus replication in K562 cells, while lovastatin could inhibit DEN-2 NGC live virus replication in human peripheral blood mononuclear cells. Renilla expression could be rescued in fluvastatin treated A549 Rluc-replicon cells after the addition of mevalonate, and partially restored with geranylgeranyl pyrophosphate, or farnesyl pyrophosphate. Our data suggest genetic and pharmacological modulation of cholesterol biosynthesis can regulate dengue virus replication.


Asunto(s)
Carboxiliasas/metabolismo , Colesterol/biosíntesis , Virus del Dengue/fisiología , ARN Interferente Pequeño/farmacología , Replicación Viral/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Carboxiliasas/genética , Virus del Dengue/efectos de los fármacos , Virus del Dengue/genética , Ácidos Grasos Monoinsaturados/farmacología , Fluvastatina , Técnicas de Silenciamiento del Gen , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Indoles/farmacología , Células K562 , Ácido Mevalónico/farmacología , Fosfatos de Poliisoprenilo/farmacología , Replicón/efectos de los fármacos , Sesquiterpenos/farmacología , Ácidos Tricarboxílicos/farmacología
8.
J Med Chem ; 52(24): 7934-7, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20014868

RESUMEN

A novel class of compounds containing N-sulfonylanthranilic acid was found to specifically inhibit dengue viral polymerase. The structural requirements for inhibition and a preliminary structure-activity relationship are described. A UV cross-linking experiment was used to map the allosteric binding site of the compound on the viral polymerase.


Asunto(s)
Virus del Dengue/enzimología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ortoaminobenzoatos/química , ortoaminobenzoatos/farmacología , Sitios de Unión , Virus del Dengue/química , Virus del Dengue/efectos de los fármacos , Inhibidores Enzimáticos/síntesis química , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , ARN Polimerasa Dependiente del ARN/química , Relación Estructura-Actividad , Ácidos Sulfínicos/síntesis química , Ácidos Sulfínicos/química , Ácidos Sulfínicos/farmacología , ortoaminobenzoatos/síntesis química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA