Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Cell ; 171(2): 273-285, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28985560

RESUMEN

Ferroptosis is a form of regulated cell death characterized by the iron-dependent accumulation of lipid hydroperoxides to lethal levels. Emerging evidence suggests that ferroptosis represents an ancient vulnerability caused by the incorporation of polyunsaturated fatty acids into cellular membranes, and cells have developed complex systems that exploit and defend against this vulnerability in different contexts. The sensitivity to ferroptosis is tightly linked to numerous biological processes, including amino acid, iron, and polyunsaturated fatty acid metabolism, and the biosynthesis of glutathione, phospholipids, NADPH, and coenzyme Q10. Ferroptosis has been implicated in the pathological cell death associated with degenerative diseases (i.e., Alzheimer's, Huntington's, and Parkinson's diseases), carcinogenesis, stroke, intracerebral hemorrhage, traumatic brain injury, ischemia-reperfusion injury, and kidney degeneration in mammals and is also implicated in heat stress in plants. Ferroptosis may also have a tumor-suppressor function that could be harnessed for cancer therapy. This Primer reviews the mechanisms underlying ferroptosis, highlights connections to other areas of biology and medicine, and recommends tools and guidelines for studying this emerging form of regulated cell death.


Asunto(s)
Muerte Celular , Animales , Apoptosis , Humanos , Hierro/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
2.
Semin Oncol ; 32(5): 465-72, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16210087

RESUMEN

This review highlights decitabine as a prototype epigenetic modifying drug to show how the clinical development of epigenetic agents differs from that of traditional cytotoxic chemotherapies. Decitabine, a cytosine analogue, is cytotoxic at high doses but has selective DNA demethylating activity at low doses. The focus of current decitabine investigations is twofold: to elucidate all of the mechanisms of action and to determine the optimal dose, schedule, and concomitant therapies. New phase I trials have identified a "biologically effective dose," which is 1 to 2 logs lower than the cytotoxic dose. A clinical development program with low-dose decitabine in malignant diseases is focused on myelodysplastic syndrome (MDS), acute myelogenous leukemia (AML), and chronic myelogenous leukemia (CML). A phase III trial in MDS showed objective responses (complete [CR] plus partial [PR] remission) and longer median time to progression to AML or death with decitabine than with supportive care alone. The optimal use of decitabine may be in combination with other agents that promote gene expression, namely, histone deacetylase (HDAC) inhibitors. Optimized decitabine doses and combinations with other epigenetic therapies that can be used at minimally toxic doses provide potentially safer therapeutic options and introduce novel combination therapies.


Asunto(s)
Azacitidina/análogos & derivados , Epigénesis Genética , Neoplasias/tratamiento farmacológico , Antígenos de Neoplasias/química , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis , Azacitidina/farmacología , Ensayos Clínicos como Asunto , Aductos de ADN , Decitabina , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Inhibidores de Histona Desacetilasas , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Neoplasias/genética , Inducción de Remisión , Factores de Tiempo , Resultado del Tratamiento
3.
Cancer ; 106(8): 1794-803, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16532500

RESUMEN

BACKGROUND: Aberrant DNA methylation, which results in leukemogenesis, is frequent in patients with myelodysplastic syndromes (MDS) and is a potential target for pharmacologic therapy. Decitabine indirectly depletes methylcytosine and causes hypomethylation of target gene promoters. METHODS: A total of 170 patients with MDS were randomized to receive either decitabine at a dose of 15 mg/m2 given intravenously over 3 hours every 8 hours for 3 days (at a dose of 135 mg/m2 per course) and repeated every 6 weeks, or best supportive care. Response was assessed using the International Working Group criteria and required that response criteria be met for at least 8 weeks. RESULTS: Patients who were treated with decitabine achieved a significantly higher overall response rate (17%), including 9% complete responses, compared with supportive care (0%) (P < .001). An additional 12 patients who were treated with decitabine (13%) achieved hematologic improvement. Responses were durable (median, 10.3 mos) and were associated with transfusion independence. Patients treated with decitabine had a trend toward a longer median time to acute myelogenous leukemia (AML) progression or death compared with patients who received supportive care alone (all patients, 12.1 mos vs. 7.8 mos [P = 0.16]; those with International Prognostic Scoring System intermediate-2/high-risk disease, 12.0 mos vs. 6.8 mos [P = 0.03]; those with de novo disease, 12.6 mos vs. 9.4 mos [P = 0.04]; and treatment-naive patients, 12.3 mos vs. 7.3 mos [P = 0.08]). CONCLUSIONS: Decitabine was found to be clinically effective in the treatment of patients with MDS, provided durable responses, and improved time to AML transformation or death. The duration of decitabine therapy may improve these results further.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Azacitidina/análogos & derivados , Síndromes Mielodisplásicos/tratamiento farmacológico , Enfermedad Aguda , Anciano , Antimetabolitos Antineoplásicos/efectos adversos , Azacitidina/administración & dosificación , Azacitidina/efectos adversos , Metilación de ADN/efectos de los fármacos , Decitabina , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Humanos , Leucemia Mieloide/etiología , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/mortalidad , Cuidados Paliativos , Calidad de Vida , Tasa de Supervivencia
4.
Blood ; 103(5): 1635-40, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14604977

RESUMEN

Decitabine (5-aza-2'-deoxycytidine) inhibits DNA methylation and has dual effects on neoplastic cells, including the reactivation of silenced genes and differentiation at low doses and cytotoxicity at high doses. We evaluated, in a phase 1 study, low-dose prolonged exposure schedules of decitabine in relapsed/refractory leukemias. Patient cohorts received decitabine at 5, 10, 15, or 20 mg/m2 intravenously over one hour daily, 5 days a week for 2 consecutive weeks, doses 5- to approximately 30-fold lower than the maximum tolerated dose (MTD). There were 2 groups that also received 15 mg/m2 daily for 15 or 20 days. A total of 50 patients were treated (44 with acute myelogenous leukemia [AML]/myelodysplasia [MDS], 5 with chronic myelogenous leukemia [CML], and 1 with acute lymphocytic leukemia [ALL]), and the drug was well tolerated at all dose levels, with myelosuppression being the major side effect. Responses were seen at all dose levels. However, the dose of 15 mg/m2 for 10 days appeared to induce the most responses (11 of 17 or 65%), with fewer responses seen when the dose was escalated or prolonged (2 of 19 or 11%). There was no correlation between P15 methylation at baseline or after therapy and response to decitabine. We conclude that decitabine is effective in myeloid malignancies, and low doses are as or more effective than higher doses.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Azacitidina/uso terapéutico , Neoplasias Hematológicas/tratamiento farmacológico , Leucemia/tratamiento farmacológico , Trastornos Mieloproliferativos/tratamiento farmacológico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Proteínas de Ciclo Celular/genética , Diferenciación Celular , Niño , Preescolar , Estudios de Cohortes , Inhibidor p15 de las Quinasas Dependientes de la Ciclina , Metilación de ADN , Metilasas de Modificación del ADN/metabolismo , Decitabina , Relación Dosis-Respuesta a Droga , Femenino , Silenciador del Gen , Humanos , Cariotipificación , Masculino , Metilación , Persona de Mediana Edad , Factores de Tiempo , Resultado del Tratamiento , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA