Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Clin Oncol ; 31(33): 4215-21, 2013 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-24127444

RESUMEN

PURPOSE: This randomized, multicenter, phase III noninferiority trial was designed to test the efficacy and safety of an oral tetra-arsenic tetra-sulfide (As4S4) -containing formula named the Realgar-Indigo naturalis formula (RIF) compared with intravenous arsenic trioxide (ATO) as both induction and maintenance therapies for newly diagnosed acute promyelocytic leukemia (APL). PATIENTS AND METHODS: In all, 242 patients with APL were randomly assigned (1:1) to oral RIF (60 mg/kg) or ATO (0.16 mg/kg) combined with all-trans retinoic acid (ATRA; 25 mg/m(2)) during induction therapy. After achieving complete remission (CR), all patients received three courses of consolidation chemotherapy and maintenance treatment with sequential ATRA followed by either RIF or ATO for 2 years. The primary end point was the rate of disease-free survival (DFS) at 2 years, which was assessed for noninferiority with a 10% noninferiority margin. RESULTS: The median follow-up time was 39 months. DFS at 2 years was 98.1% (106 of 108) in the RIF group and 95.5% (107 of 112) in the ATO group. The DFS difference was 2.6% (95% CI, -3.0% to 8.0%). The lower limit of the 95% CI of DFS difference was greater than the -10% noninferiority margin, confirming noninferiority (P < .001). No significant differences were noted between the RIF and ATO groups with regard to the CR rate (99.1% v 97.2%; P = .62) or the overall survival at 3 years (99.1% v 96.6%; P = .18). The rates of adverse events were similar in the two groups. CONCLUSION: Oral RIF plus ATRA is not inferior to intravenous ATO plus ATRA as first-line treatment of APL and may be considered as a routine treatment option for appropriate patients.


Asunto(s)
Arsenicales/uso terapéutico , Medicamentos Herbarios Chinos/uso terapéutico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Óxidos/uso terapéutico , Sulfuros/uso terapéutico , Administración Intravenosa , Administración Oral , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Trióxido de Arsénico , Arsenicales/administración & dosificación , Arsenicales/efectos adversos , Supervivencia sin Enfermedad , Medicamentos Herbarios Chinos/administración & dosificación , Femenino , Fiebre/inducido químicamente , Estudios de Seguimiento , Humanos , Quimioterapia de Inducción/métodos , Hígado/efectos de los fármacos , Hígado/patología , Quimioterapia de Mantención/métodos , Masculino , Persona de Mediana Edad , Óxidos/administración & dosificación , Óxidos/efectos adversos , Inducción de Remisión , Sulfuros/administración & dosificación , Resultado del Tratamiento , Tretinoina/administración & dosificación , Tretinoina/uso terapéutico , Adulto Joven
2.
Proc Natl Acad Sci U S A ; 110(6): 2258-63, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23345445

RESUMEN

Eriocalyxin B (EriB), a diterpenoid isolated from Isodon eriocalyx, was previously reported to have antitumor effects via multiple pathways, and these pathways are related to immune responses. In this study, we demonstrated that EriB was efficacious in experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis. Treatment with EriB led to amelioration of EAE, which correlated with reduced spinal cord inflammation and demyelination. EriB treatment abolished encephalitogenic T-cell responses to myelin oligodendrocyte glycoprotein in an adoptive transfer EAE model. The underlying mechanism of EriB-induced effects involved inhibition of T helper (Th) 1 and Th17 cell differentiation through Janus Kinase/Signal Transducer and Activator Of Transcription and Nuclear factor-κB signaling pathways as well as elevation of reactive oxygen species. These findings indicate that EriB exerts potent antiinflammatory effects through selective modulation of pathogenic Th1 and Th17 cells by targeting critical signaling pathways. The study provides insights into the role of EriB as a unique therapeutic agent for the treatment of autoimmune diseases.


Asunto(s)
Diterpenos/farmacología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Células TH1/efectos de los fármacos , Células Th17/efectos de los fármacos , Traslado Adoptivo , Animales , Autoinmunidad/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Inmunosupresores/farmacología , Medicina Tradicional China , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Células TH1/inmunología , Células Th17/inmunología
3.
Blood ; 117(24): 6425-37, 2011 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-21422471

RESUMEN

Arsenic had been used in treating malignancies from the 18th to mid-20th century. In the past 3 decades, arsenic was revived and shown to be able to induce complete remission and to achieve, when combined with all-trans retinoic acid and chemotherapy, a 5-year overall survival of 90% in patients with acute promyelocytic leukemia driven by the t(15;17) translocation-generated promyelocytic leukemia-retinoic acid receptor α (PML-RARα) fusion. Molecularly, arsenic binds thiol residues and induces the formation of reactive oxygen species, thus affecting numerous signaling pathways. Interestingly, arsenic directly binds the C3HC4 zinc finger motif in the RBCC domain of PML and PML-RARα, induces their homodimerization and multimerization, and enhances their interaction with the SUMO E2 conjugase Ubc9, facilitating subsequent sumoylation/ubiquitination and proteasomal degradation. Arsenic-caused intermolecular disulfide formation in PML also contributes to PML-multimerization. All-trans retinoic acid, which targets PML-RARα for degradation through its RARα moiety, synergizes with arsenic in eliminating leukemia-initiating cells. Arsenic perturbs a number of proteins involved in other hematologic malignancies, including chronic myeloid leukemia and adult T-cell leukemia/lymphoma, whereby it may bring new therapeutic benefits. The successful revival of arsenic in acute promyelocytic leukemia, together with modern mechanistic studies, has thus allowed a new paradigm to emerge in translational medicine.


Asunto(s)
Arsénico/uso terapéutico , Leucemia/tratamiento farmacológico , Leucemia/genética , Medicina Tradicional/métodos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Adulto , Animales , Arsénico/farmacología , Humanos , Magia , Medicina Tradicional/tendencias , Modelos Biológicos , Modelos Moleculares , Resultado del Tratamiento
4.
Nat Genet ; 43(4): 309-15, 2011 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-21399634

RESUMEN

Abnormal epigenetic regulation has been implicated in oncogenesis. We report here the identification of somatic mutations by exome sequencing in acute monocytic leukemia, the M5 subtype of acute myeloid leukemia (AML-M5). We discovered mutations in DNMT3A (encoding DNA methyltransferase 3A) in 23 of 112 (20.5%) cases. The DNMT3A mutants showed reduced enzymatic activity or aberrant affinity to histone H3 in vitro. Notably, there were alterations of DNA methylation patterns and/or gene expression profiles (such as HOXB genes) in samples with DNMT3A mutations as compared with those without such changes. Leukemias with DNMT3A mutations constituted a group of poor prognosis with elderly disease onset and of promonocytic as well as monocytic predominance among AML-M5 individuals. Screening other leukemia subtypes showed Arg882 alterations in 13.6% of acute myelomonocytic leukemia (AML-M4) cases. Our work suggests a contribution of aberrant DNA methyltransferase activity to the pathogenesis of acute monocytic leukemia and provides a useful new biomarker for relevant cases.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Leucemia Monocítica Aguda/enzimología , Leucemia Monocítica Aguda/genética , Mutación Missense , Adulto , Anciano , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Secuencia de Bases , Biomarcadores de Tumor/genética , Secuencia Conservada , ADN (Citosina-5-)-Metiltransferasas/química , Metilación de ADN/genética , ADN Metiltransferasa 3A , ADN Complementario/genética , Exones , Femenino , N-Metiltransferasa de Histona-Lisina , Humanos , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/genética , Masculino , Persona de Mediana Edad , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Pronóstico , Homología de Secuencia de Aminoácido
5.
Science ; 328(5975): 240-3, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20378816

RESUMEN

Arsenic, an ancient drug used in traditional Chinese medicine, has attracted worldwide interest because it shows substantial anticancer activity in patients with acute promyelocytic leukemia (APL). Arsenic trioxide (As2O3) exerts its therapeutic effect by promoting degradation of an oncogenic protein that drives the growth of APL cells, PML-RARalpha (a fusion protein containing sequences from the PML zinc finger protein and retinoic acid receptor alpha). PML and PML-RARalpha degradation is triggered by their SUMOylation, but the mechanism by which As2O3 induces this posttranslational modification is unclear. Here we show that arsenic binds directly to cysteine residues in zinc fingers located within the RBCC domain of PML-RARalpha and PML. Arsenic binding induces PML oligomerization, which increases its interaction with the small ubiquitin-like protein modifier (SUMO)-conjugating enzyme UBC9, resulting in enhanced SUMOylation and degradation. The identification of PML as a direct target of As2O3 provides new insights into the drug's mechanism of action and its specificity for APL.


Asunto(s)
Arsénico/metabolismo , Arsenicales/metabolismo , Arsenicales/farmacología , Proteínas Nucleares/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Óxidos/metabolismo , Óxidos/farmacología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Trióxido de Arsénico , Línea Celular , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/genética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Oxazinas/metabolismo , Proteína de la Leucemia Promielocítica , Conformación Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de Ácido Retinoico/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Receptor alfa de Ácido Retinoico , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Factores de Transcripción/química , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética , Ubiquitinación , Dedos de Zinc
7.
Proc Natl Acad Sci U S A ; 105(12): 4826-31, 2008 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-18344322

RESUMEN

To enhance therapeutic efficacy and reduce adverse effects, practitioners of traditional Chinese medicine (TCM) prescribe a combination of plant species/minerals, called formulae, based on clinical experience. Nearly 100,000 formulae have been recorded, but the working mechanisms of most remain unknown. In trying to address the possible beneficial effects of formulae with current biomedical approaches, we use Realgar-Indigo naturalis formula (RIF), which has been proven to be very effective in treating human acute promyelocytic leukemia (APL) as a model. The main components of RIF are realgar, Indigo naturalis, and Salvia miltiorrhiza, with tetraarsenic tetrasulfide (A), indirubin (I), and tanshinone IIA (T) as major active ingredients, respectively. Here, we report that the ATI combination yields synergy in the treatment of a murine APL model in vivo and in the induction of APL cell differentiation in vitro. ATI causes intensified ubiquitination/degradation of promyelocytic leukemia (PML)-retinoic acid receptor alpha (RARalpha) oncoprotein, stronger reprogramming of myeloid differentiation regulators, and enhanced G(1)/G(0) arrest in APL cells through hitting multiple targets compared with the effects of mono- or biagents. Furthermore, ATI intensifies the expression of Aquaglyceroporin 9 and facilitates the transportation of A into APL cells, which in turn enhances A-mediated PML-RARalpha degradation and therapeutic efficacy. Our data also indicate A as the principal component of the formula, whereas T and I serve as adjuvant ingredients. We therefore suggest that dissecting the mode of action of clinically effective formulae at the molecular, cellular, and organism levels may be a good strategy in exploring the value of traditional medicine.


Asunto(s)
Arsenicales/uso terapéutico , Medicamentos Herbarios Chinos/uso terapéutico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Medicina Tradicional China , Sulfuros/uso terapéutico , Animales , Acuaporinas/genética , Acuaporinas/metabolismo , Arsenicales/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Medicamentos Herbarios Chinos/farmacología , Fase G1/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patología , Ratones , Proteínas de Fusión Oncogénica/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Sulfuros/farmacología , Transcripción Genética/efectos de los fármacos , Ubiquitinación/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
9.
Blood ; 109(8): 3441-50, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17197433

RESUMEN

Studies have documented the potential antitumor activities of oridonin, a compound extracted from medicinal herbs. However, whether oridonin can be used in the selected setting of hematology/oncology remains obscure. Here, we reported that oridonin induced apoptosis of t(8;21) acute myeloid leukemic (AML) cells. Intriguingly, the t(8;21) product AML1-ETO (AE) fusion protein, which plays a critical role in leukemogenesis, was degraded with generation of a catabolic fragment, while the expression pattern of AE target genes investigated could be reprogrammed. The ectopic expression of AE enhanced the apoptotic effect of oridonin in U937 cells. Preincubation with caspase inhibitors blocked oridonin-triggered cleavage of AE, while substitution of Ala for Asp at residues 188 in ETO moiety of the fusion abrogated AE degradation. Furthermore, oridonin prolonged lifespan of C57 mice bearing truncated AE-expressing leukemic cells without suppression of bone marrow or reduction of body weight of animals, and exerted synergic effects while combined with cytosine arabinoside. Oridonin also inhibited tumor growth in nude mice inoculated with t(8;21)-harboring Kasumi-1 cells. These results suggest that oridonin may be a potential antileukemia agent that targets AE oncoprotein at residue D188 with low adverse effect, and may be helpful for the treatment of patients with t(8;21) AML.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Subunidad alfa 2 del Factor de Unión al Sitio Principal/antagonistas & inhibidores , Diterpenos de Tipo Kaurano/farmacología , Diterpenos/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Animales , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 8/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Citarabina/agonistas , Citarabina/farmacología , Diterpenos/agonistas , Diterpenos/química , Diterpenos de Tipo Kaurano/agonistas , Diterpenos de Tipo Kaurano/química , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Desnudos , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Proteínas de Fusión Oncogénica/metabolismo , Extractos Vegetales/agonistas , Extractos Vegetales/química , Extractos Vegetales/farmacología , Plantas Medicinales/química , Proteína 1 Compañera de Translocación de RUNX1 , Translocación Genética , Células U937
10.
Cancer Biol Ther ; 1(6): 614-20, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12642682

RESUMEN

Most acute promyelocytic leukemia (APL) cases have t(15;17)(q22;q21) chromosomal translocation and PML-RARalpha chimeric gene which blocks granulocytic differentiation. The introduction of all-trans-retinoic acid (ATRA) and arsenic compounds, especially arsenic trioxide (As(2)O(3)), has provided good models to study not only differentiation and/or apoptosis therapy but also molecular target-based cancer treatment. In vivo and in vitro investigations have shown that both agents are able to induce differentiation of APL cells: ATRA tends to induce terminal differentiation, while low-dose As(2)O(3) can induce partial differentiation. Significant progress has been made in understanding the molecular mechanisms of APL pathogenesis and differentiation therapy. Pharmacological concentrations (0.1 approximately 1 microM) of ATRA derepresses transcription by releasing CoR from, and recruiting CoA to PML-RARalpha, whereas As(2)O(3) triggers a rapid degradation of PML-RARalpha. In fact, the two drugs act on the same oncoprotein through targeting different moieties and in distinct ways and thereby abrogate its dominant-negative effects on regulatory pathways necessary for granulocytic differentiation. As to apoptosis, it is clear that high-dose As(2)O(3) can induce mitochondria-mediated cell death pathway in a thiol-dependent manner, while the mechanism of ATRA-induced apoptosis needs further elucidation. Transcriptomic and proteomic analysis are also expected to find new molecular targets. It is the hope that what we have learnt from APL will benefit further developments of anti-leukemia therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Arsenicales/uso terapéutico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Óxidos/uso terapéutico , Tretinoina/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Diferenciación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Leucemia Promielocítica Aguda/patología , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Fusión Oncogénica/efectos de los fármacos , Translocación Genética/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA