Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 68(14): 5849-58, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18632639

RESUMEN

Tissue type transglutaminase (TG2) is a unique multifunctional protein that plays a role in many steps in the cancer metastatic cascade. Here, we examined the clinical (n = 93 epithelial ovarian cancers) and biological (in vitro adhesion, invasion, and survival and in vivo therapeutic targeting) significance of TG2 in ovarian cancer. The overexpression of TG2 was associated with significantly worse overall patient survival in both univariate and multivariate analyses. Transfection of TG2 into SKOV3ip1 cells promoted attachment and spreading on fibronectin-coated surfaces and increased the in vitro invasive potential of these cells. Conversely, TG2 silencing with small interfering RNA (siRNA) of HeyA8 cells significantly decreased the invasive potential of the cells and also increased docetaxel-induced cell death. In vivo therapy experiments using chemotherapy-sensitive (HeyA8) and chemotherapy-resistant (HeyA8-MDR and RMG2) models showed significant antitumor activity both with TG2 siRNA-1,2-dioleoyl-sn-glycero-3-phosphatidylcholine alone and in combination with docetaxel chemotherapy. This antitumor activity was related to decreased proliferation and angiogenesis and increased tumor cell apoptosis in vivo. Taken together, these findings indicate that TG2 overexpression is an adverse prognostic factor in ovarian carcinoma and TG2 targeting may be an attractive therapeutic approach.


Asunto(s)
Carcinoma/enzimología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/enzimología , Transglutaminasas/biosíntesis , Animales , Antineoplásicos/farmacología , Carcinoma/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Proteínas de Unión al GTP , Silenciador del Gen , Humanos , Ratones , Mutación , Trasplante de Neoplasias , Neoplasias Ováricas/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2
2.
Mol Cancer Ther ; 5(6): 1593-601, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16818519

RESUMEN

The antiapoptotic protein Bcl-2 is overexpressed in a majority of breast cancers, and is associated with a diminished apoptotic response and resistance to various antitumor agents. Bcl-2 inhibition is currently being explored as a possible strategy for sensitizing breast cancer cells to standard chemotherapeutic agents. Antisense Bcl-2 oligonucleotides represent one method for blocking the antiapoptotic effects of Bcl-2. In this study, we show that antisense Bcl-2 efficiently blocks Bcl-2 expression, resulting in the apoptosis of breast cancer cells. Antisense Bcl-2-mediated cytotoxicity was associated with the induction of the B cell translocation gene 1 (BTG1). Importantly, knockdown of BTG1 reduced antisense Bcl-2-mediated cytotoxicity in breast cancer cells. Furthermore, BTG1 expression seems to be negatively regulated by Bcl-2, and exogenous expression of BTG1 induced apoptosis. These results suggest that BTG1 is a Bcl-2-regulated mediator of apoptosis in breast cancer cells, and that its induction contributes to antisense Bcl-2-mediated cytotoxic effects.


Asunto(s)
Apoptosis , Neoplasias de la Mama/patología , Proteínas de Neoplasias/metabolismo , Oligonucleótidos Antisentido/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Humanos , Leucemia Linfocítica Crónica de Células B/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/genética , Células Tumorales Cultivadas
3.
Cancer ; 100(7): 1365-73, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15042669

RESUMEN

BACKGROUND: Complete pathologic response of breast carcinoma to neoadjuvant chemotherapy is a well defined outcome that correlates with prolonged survival. Categorization of incomplete response depends on accurate measurement of residual tumor size but is complicated by the variable histopathologic changes that occur within the tumor bed. In the current study, the authors investigated the contribution of assessing tumor cellularity in the pathologic evaluation of response to chemotherapy. METHODS: The slides from diagnostic core needle biopsy and the subsequent matched resection specimens were examined in 240 patients with breast carcinoma: 120 "treated" patients who received neoadjuvant chemotherapy and 120 "control" patients who received primary surgical management within a few weeks of diagnosis. Clinical response and residual tumor size were evaluated in 108 treated patients who completed a clinical trial with paclitaxel and then received combined 5-fluorouracil, doxorubicin, and cyclophosphamide chemotherapy. Tumor cellularity was assessed from hematoxylin and eosin-stained tissue sections as the percentage of tumor area that contained invasive carcinoma. RESULTS: After neoadjuvant chemotherapy, tumor cellularity decreased from a median of 40% in core needle biopsy to 10% in resection specimens (P<0.01; Wilcoxon signed rank test). The cellularity of core needle biopsy (median, 30%) tended to underestimate the cellularity of resection specimens (median, 40%) in the control group (P<0.01). Changes in cellularity varied within each clinical response category, particularly partial response and minor response. The greatest reduction was observed in the cellularity of residual primary tumors that measured < or =1 cm (pathologic T1a [pT1a] and pT1b tumors), but changes in cellularity varied in the pT1, pT2, and pT3 residual tumor categories. The shape of the distribution of tumor size, expressed as the greatest dimension in cm, was similar in the control group and the treatment group (excluding complete pathologic response); however, when residual tumor size and cellularity were combined, the distribution of pathologic response shifted left (toward complete response) with a steep decline, suggesting that many tumors had a large reduction in cellularity but little change in the tumor size. CONCLUSIONS: Cellularity of the tumor mass was reduced significantly by neoadjuvant chemotherapy, and the change varied widely in different categories of clinical response. Although residual tumors measuring < or =1 cm in greatest dimension had the most reduction in tumor cellularity, there was broad variability for all residual tumor groups (pT1-pT3). The frequency distribution of residual tumor size was altered markedly by the inclusion of tumor cellularity, indicating that the product of pathologic size and tumor cellularity may provide more accurate pathologic response information than tumor size alone.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Ciclofosfamida/uso terapéutico , Doxorrubicina/uso terapéutico , Fluorouracilo/uso terapéutico , Paclitaxel/uso terapéutico , Biopsia con Aguja , Neoplasias de la Mama/cirugía , Quimioterapia Adyuvante , Femenino , Humanos , Terapia Neoadyuvante , Neoplasia Residual , Resultado del Tratamiento
4.
Cancer Chemother Pharmacol ; 53(3): 191-203, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14614574

RESUMEN

PURPOSE: Structure-activity studies were carried out with the model bioreductive alkylating agent benzoquinone mustard (BM) and its structural analogs. The specific objectives were: (1) to investigate the effects of functional group substitutions to the benzoquinone ring on DNA crosslink and strand break formation subsequent to reduction of the analogs by DT-diaphorase (DTD) in vitro, (2) to correlate DNA crosslink and strand break formation by the analogs with anaerobic reduction of the BM analogs by DTD and their redox cycling in vitro, and (3) to correlate DNA crosslink and strand break formation by the BM analogs with their cytotoxic effects in cancer cells. METHODS: DNA interstrand crosslink and single-strand break formation were assessed using agarose gel assays. To determine DNA interstrand crosslinks or single-strand breaks, linearized or supercoiled plasmid DNA, respectively, were incubated with purified human DTD and increasing concentrations of each BM analog. Subsequently, DNA was electrophoresed on an agarose gel and DNA crosslink and strand break formation were quantified using densitometry. The rates of reduction of the BM analogs by purified human DTD were measured in vitro under hypoxic conditions, and the redox cycling potential was determined under aerobic conditions using HPLC analysis. The cytotoxic activities of these agents in human tumor cell lines were measured by the MTT assay, with and without the DTD inhibitor, dicoumarol. RESULTS: BM analogs with electron-donating groups (MeBM, MBM, m-MeBM), electron-withdrawing groups (CBM, FBM), sterically bulky groups (PBM, m-PBM, m-TBM) and positional isomers (MeBM, m-MeBM, PBM, m-PBM) were synthesized. After reduction by DTD, the BM analogs produced a concentration-dependent increase in DNA crosslink and DNA strand break formation. The E(10) (extent of DNA crosslink formation produced by 10 micro M BM analog) for DNA crosslink formation displayed the rank order MeBM approximately MBM>m-MeBM approximately PBM approximately BM>CBM>FBM>m-PBM approximately m-TBM. For DNA strand break formation, the E(10) values (extent of DNA strand break formation produced by 10 micro M BM analog) displayed the rank order MeBM>MBM>m-MeBM>PBM>BM approximately CBM>FBM>m-PBM approximately m-TBM. Importantly, the cytotoxic activity of the BM analogs in SK-Mel-28 human melanoma cells correlated positively with the E(10) values for DTD-mediated DNA crosslink formation ( r(s)=0.87, P<0.05) and DNA strand break formation ( r(s)=0.95, P<0.05). Similar correlations were observed in NCI-H661 human lung carcinoma cells. Furthermore, the D(10) values (concentration of BM analog that decreased the surviving cell fraction to 0.1) for cytotoxic activity of the BM analogs correlated with the maximum levels of DNA crosslinks formed with each BM analog, with r(s) values of -0.85 ( P<0.05) for the NCI-H661 cell line, and -0.81 ( P<0.05) for the SK-MEL-28 cell line. The half-time of reduction (t(1/2)) of the BM analogs by DTD did not correlate with DNA crosslink formation, DNA strand break formation, or cytotoxic potency of the analogs. CONCLUSIONS: Functional groups on the benzoquinone ring affect the ability of BM to produce DNA crosslinks and strand breaks following reduction by DTD. Electron-donating groups increased DNA damage, whereas electron-withdrawing groups and sterically bulky groups at the C6 position had no effect or decreased the ability of the compounds to produce DNA damage compared to BM. Moreover, both DNA crosslink and strand break formation appear to have an important impact on the cytotoxicity of the BM analogs. These results may have significance for optimal use of BM-based antitumor agents and for rationalization of the development of novel therapeutic compounds that require bioactivation by DTD.


Asunto(s)
Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/toxicidad , Benzoquinonas/química , Benzoquinonas/toxicidad , Daño del ADN , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Antineoplásicos Alquilantes/metabolismo , Benzoquinonas/metabolismo , Línea Celular Tumoral , Reactivos de Enlaces Cruzados/química , Reactivos de Enlaces Cruzados/metabolismo , Reactivos de Enlaces Cruzados/toxicidad , ADN/análisis , Relación Dosis-Respuesta a Droga , Humanos , Oxidación-Reducción , Relación Estructura-Actividad
5.
Cancer Chemother Pharmacol ; 49(2): 101-10, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11862423

RESUMEN

PURPOSE: Bioreductive antitumor agents are an important class of anticancer drugs that include the clinically used drug, mitomycin C, and new agents such as EO9 and tirapazamine that have recently been tested in clinical trials. These agents require activation by reductive enzymes such as DT-diaphorase or NADPH:cytochrome P450 reductase. A major focus for improving cancer chemotherapy has been to increase the selectivity and targeting of antitumor drugs to tumor cells. Bioreductive antitumor agents are ideally suited to improving tumor selectivity by an enzyme-directed approach to tumor targeting. However, none of the bioreductive agents developed to date has been specific for activation by a single reductive enzyme. This is in part due to a lack of knowledge about structural factors that confer selectivity for activation by reductive enzymes. The purpose of this study was to investigate the ability of specific functional groups to modify reduction and activation of quinone bioreductive agents by DT-diaphorase. METHODS: We used a series of model benzoquinone mustard (BM) bioreductive agents and compared the parent compound BM to MBM, which has a strong electron-donating methoxy group, MeBM, which has a weaker electron-donating methyl group, CBM, which has an electron-withdrawing chloro group, and PBM and its structural isomer, meta-PBM (m-PBM), which both have sterically bulky benzene rings attached to the quinone moiety. We determined the rate of reduction of these agents by purified human DT-diaphorase under hypoxic and aerobic conditions. We also measured the cytotoxic activity of these agents in human tumor cell lines with and without the DT-diaphorase inhibitor, dicoumarol. RESULTS: Under hypoxic conditions in vitro, the t(1/2) values for reduction of the analogs by purified DT-diaphorase were 4, 6, 8, 9, 10 and 21 min for BM, MeBM, CBM, MBM, PBM and m-PBM, respectively. Under aerobic conditions the rank order of redox cycling after two-electron reduction by DT-diaphorase was MBM > MeBM > BM approximately CBM approximately PBM approximately m-PBM. The rate of reduction by DT-diaphorase of HBM, a non-alkylating analog of BM, was similar to that of BM under hypoxic conditions, and the rate of redox cycling under aerobic conditions was comparable to that of BM, suggesting that structural changes to the cytotoxic group of these BMs do not affect DT-diaphorase-mediated reduction and redox cycling potential. MBM, MeBM and PBM were more toxic than BM in the NCI-H661 human non-small-cell lung cancer cells and SK-MEL-28 human melanoma cells, while CBM displayed significantly increased cytotoxic activity compared to BM only in the NCI H661 cells. m-PBM had similar cytotoxic activity compared with BM in both cell lines. These cell lines have moderate to high levels of DT-diaphorase activity. When cells were pretreated with the DT-diaphorase inhibitor, dicoumarol, the cytotoxic activity of BM increased while that of MBM decreased in both cell lines, suggesting that BM was inactivated by DT-diaphorase while MBM was activated by this enzyme. Pretreatment of the SK-MEL-28 melanoma cells with dicoumarol resulted in an increased cytotoxic activity of MeBM, but pretreatment of the NCI-H661 cells did not affect the cytotoxicity of MeBM. This suggests, that similar to the results with BM, DT-diaphorase is an inactivating enzyme for MeBM in the SK-MEL-28 cell line. Dicoumarol had no significant effect on the cytotoxicity of CBM, PBM or m-PBM in both cell lines. CONCLUSIONS: These studies demonstrated that functional groups can significantly affect the reduction and activation of bioreductive agents by DT-diaphorase. All the functional groups decreased the rate of reduction of the quinone group by DT-diaphorase. Since MeBM and MBM, with electron-donating functional groups, and CBM with an electron-withdrawing functional group had similar half-lives of reduction by DT-diaphorase, steric rather than electronic effects of the functional groups appear to be more important for modifying the rate of reduction by DT-diaphorase. Steric effects on reduction by DT-diaphorase were also influenced by the position of the functional group on the quinone ring moiety, as the reduction of m-PBM was much slower than the reduction of PBM. The electron-donating methoxy and methyl functional groups increased the ability of the reduced products of MBM and MeBM to undergo redox cycling. DT-diaphorase appeared to be an activating enzyme for MBM. This may have resulted in part from increased formation of reactive oxygen species resulting from the increased redox cycling by MBM. In contrast, DT-diaphorase was an inactivating enzyme for BM, and for MeBM in the SK-MEL-28 melanoma cells, possibly because the hydroquinone product of BM and MeBM may be less cytotoxic than the semiquinone produced by one-electron reduction by NADPH:cytochrome P450 reductase.


Asunto(s)
Antineoplásicos/metabolismo , Benzoquinonas/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Antineoplásicos/farmacología , Benzoquinonas/farmacología , Biotransformación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , NAD/metabolismo , Oxidación-Reducción , Relación Estructura-Actividad , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...