Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
FASEB J ; 35(10): e21885, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34478585

RESUMEN

In a recently published phase III clinical trial, gemcitabine (GEM) plus cisplatin (DDP) induction chemotherapy significantly improved recurrence-free survival and overall survival and became the standard of care among patients with locoregionally advanced NPC. However, the molecular mechanisms of GEM synergized with DPP in NPC cells remain elucidated. These findings prompt us to explore the effect of the combination between GEM and DDP in NPC cell lines through proliferative phenotype, immunofluorescence, flow cytometry, and western blotting assays. In vitro studies reveal that GEM or DPP treated alone induces cell cycle arrest, promotes cell apoptosis, forces DNA damage response, and GEM synergism with DDP significantly increases the above effects in NPC cells. In vivo studies indicate that GEM or DPP treated alone significantly inhibits the tumor growth and prolongs the survival time of mice injected with SUNE1 cells compared to the control group. Moreover, the mice treated with GEM combined with DDP have smaller tumors and survive longer than those in GEM or DPP treated alone group. In addition, P-gp may be the key molecule that regulates the synergistic effect of gemcitabine and cisplatin. GEM synergizes with DPP to inhibit NPC cell proliferation and tumor growth by inducing cell cycle arrest, cell apoptosis, and DNA damage response, which reveals the mechanisms of combined GEM and DDP induction chemotherapy in improving locoregionally advanced NPC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proliferación Celular/efectos de los fármacos , Carcinoma Nasofaríngeo/tratamiento farmacológico , Neoplasias Nasofaríngeas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Cisplatino/agonistas , Cisplatino/farmacología , Desoxicitidina/agonistas , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Sinergismo Farmacológico , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
2.
PLoS One ; 8(3): e57523, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23520471

RESUMEN

Sarcomas are rare and heterogeneous mesenchymal tumors affecting both pediatric and adult populations with more than 70 recognized histologies. Doxorubicin and ifosfamide have been the main course of therapy for treatment of sarcomas; however, the response rate to these therapies is about 10-20% in metastatic setting. Toxicity with the drug combination is high, response rates remain low, and improvement in overall survival, especially in the metastatic disease, remains negligible and new agents are needed. Wee1 is a critical component of the G2/M cell cycle checkpoint control and mediates cell cycle arrest by regulating the phosphorylation of CDC2. Inhibition of Wee1 by MK1775 has been reported to enhance the cytotoxic effect of DNA damaging agents in different types of carcinomas. In this study we investigated the therapeutic efficacy of MK1775 in various sarcoma cell lines, patient-derived tumor explants ex vivo and in vivo both alone and in combination with gemcitabine, which is frequently used in the treatment of sarcomas. Our data demonstrate that MK1775 treatment as a single agent at clinically relevant concentrations leads to unscheduled entry into mitosis and initiation of apoptotic cell death in all sarcomas tested. Additionally, MK1775 significantly enhances the cytotoxic effect of gemcitabine in sarcoma cells lines with different p53 mutational status. In patient-derived bone and soft tissue sarcoma samples we showed that MK1775 alone and in combination with gemcitabine causes significant apoptotic cell death. Magnetic resonance imaging (MRI) and histopathologic studies showed that MK1775 induces significant cell death and terminal differentiation in a patient-derived xenograft mouse model of osteosarcoma in vivo. Our results together with the high safety profile of MK1775 strongly suggest that this drug can be used as a potential therapeutic agent in the treatment of both adult as well as pediatric sarcoma patients.


Asunto(s)
Antimetabolitos Antineoplásicos , Proteínas de Ciclo Celular/antagonistas & inhibidores , Desoxicitidina/análogos & derivados , Neoplasias Femorales/tratamiento farmacológico , Proteínas Nucleares/antagonistas & inhibidores , Osteosarcoma/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles , Pirimidinas , Adolescente , Adulto , Animales , Antimetabolitos Antineoplásicos/agonistas , Antimetabolitos Antineoplásicos/farmacología , Muerte Celular , Diferenciación Celular , Línea Celular Tumoral , Niño , Preescolar , Desoxicitidina/agonistas , Desoxicitidina/farmacología , Sinergismo Farmacológico , Femenino , Neoplasias Femorales/patología , Humanos , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Trasplante de Neoplasias , Osteosarcoma/patología , Pirazoles/agonistas , Pirazoles/farmacología , Pirimidinas/agonistas , Pirimidinas/farmacología , Pirimidinonas , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
3.
Exp Hematol ; 40(10): 800-10, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22687754

RESUMEN

Hematopoietic stem cell transplantation is used for treatment of lymphoma. In an attempt to design an efficacious and safe prehematopoietic stem cell transplantation conditioning regimen, we investigated the cytotoxicity of the combination of busulfan (B), melphalan (M), and gemcitabine (G) in lymphoma cell lines in the absence or presence of drugs that induce epigenetic changes. Cells were exposed to drugs individually or in combination and analyzed by the MTT proliferation assay, flow cytometry, and Western blotting. We used ~IC(10) drug concentrations (57 µM B, 1 µM M and 0.02 µM G), which individually did not have major effects on cell proliferation. Their combination resulted in 50% inhibition of proliferation. Reduction to almost half concentration (20 µM B, 0.7 µM M and 0.01 µM G) did not have significant effects, but addition of the histone deacetylase inhibitor suberoylanilide hydroxamic acid (0.6 µM) to this combination resulted in a marked (~65%) growth inhibition. The cytotoxicity of these combinations correlates with the activation of the ataxia telangiectasia mutated-CHK2 pathway, phosphorylation of KRAB-associated protein-1, epigenetic changes such as methylation and acetylation of histone 3, and activation of apoptosis. The relevance of epigenetic changes is further shown by the induction of DNA methyltransferases in tumor cells with low constitutive levels of DNMT3A and DNMT3B. The addition of 5-aza-2'-deoxycytidine to (BMG+suberoylanilide hydroxamic acid) further enhances cell killing. Overall, BMG combinations are synergistically cytotoxic to lymphoma cells. Epigenetic changes induced by suberoylanilide hydroxamic acid and 5-aza-2'-deoxycytidine further enhance the cytotoxicity. This study provides a rationale for an ongoing clinical trial in our institution using (BMG+suberoylanilide hydroxamic acid) as pre-hematopoietic stem cell transplantation conditioning for lymphoma.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Azacitidina/análogos & derivados , Proliferación Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Epigénesis Genética/efectos de los fármacos , Linfoma/metabolismo , Linfoma/terapia , Antineoplásicos Alquilantes/agonistas , Proteínas de la Ataxia Telangiectasia Mutada , Azacitidina/agonistas , Azacitidina/farmacología , Busulfano/agonistas , Busulfano/farmacología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Quinasa de Punto de Control 2 , Citotoxinas , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Proteínas de Unión al ADN/metabolismo , Decitabina , Desoxicitidina/agonistas , Desoxicitidina/farmacología , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales/métodos , Sinergismo Farmacológico , Trasplante de Células Madre Hematopoyéticas , Humanos , Ácidos Hidroxámicos/agonistas , Ácidos Hidroxámicos/farmacología , Linfoma/patología , Melfalán/agonistas , Melfalán/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo , Proteínas Supresoras de Tumor/metabolismo , Gemcitabina , ADN Metiltransferasa 3B
4.
Blood ; 118(15): 4140-9, 2011 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-21844567

RESUMEN

Mantle cell lymphoma (MCL) usually responds well to initial therapy but is prone to relapses with chemoresistant disease, indicating the need for novel therapeutic approaches. Inhibition of the p53 E3 ligase human homolog of the murine double minute protein-2 (HDM-2) with MI-63 has been validated as one such strategy in wild-type (wt) p53 models, and our genomic and proteomic analyses demonstrated that MI-63 suppressed the expression of the ribonucleotide reductase (RNR) subunit M2 (RRM2). This effect occurred in association with induction of p21 and cell-cycle arrest at G(1)/S and prompted us to examine combinations with the RNR inhibitor 2',2'-difluoro-2'-deoxycytidine (gemcitabine). The regimen of MI-63-gemcitabine induced enhanced, synergistic antiproliferative, and proapoptotic effects in wtp53 MCL cell lines. Addition of exogenous dNTPs reversed this effect, whereas shRNA-mediated inhibition of RRM2 was sufficient to induce synergy with gemcitabine. Combination therapy of MCL murine xenografts with gemcitabine and MI-219, the in vivo analog of MI-63, resulted in enhanced antitumor activity. Finally, synergy was seen with MI-63-gemcitabine in primary patient samples that were found to express high levels of RRM2 compared with MCL cell lines. These findings provide a framework for translation of the rational combination of an HDM-2 and RNR inhibitor to the clinic for patients with relapsed wtp53 MCL.


Asunto(s)
Antimetabolitos Antineoplásicos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Desoxicitidina/análogos & derivados , Indoles , Linfoma de Células del Manto/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Ribonucleósido Difosfato Reductasa/biosíntesis , Compuestos de Espiro , Animales , Antimetabolitos Antineoplásicos/agonistas , Antimetabolitos Antineoplásicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Desoxicitidina/agonistas , Desoxicitidina/farmacología , Sinergismo Farmacológico , Fase G1/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/agonistas , Indoles/farmacología , Linfoma de Células del Manto/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Fase S/efectos de los fármacos , Compuestos de Espiro/agonistas , Compuestos de Espiro/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA