Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
N Engl J Med ; 358(18): 1919-28, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18450603

RESUMEN

BACKGROUND: A role of microRNAs in cancer has recently been recognized. However, little is known about the role of microRNAs in acute myeloid leukemia (AML). METHODS: Using microRNA expression profiling, we studied samples of leukemia cells from adults under the age of 60 years who had cytogenetically normal AML and high-risk molecular features--that is, an internal tandem duplication in the fms-related tyrosine kinase 3 gene (FLT3-ITD), a wild-type nucleophosmin (NPM1), or both. A microRNA signature that was associated with event-free survival was derived from a training group of 64 patients and tested in a validation group of 55 patients. For the latter, a microRNA compound covariate predictor (called a microRNA summary value) was computed on the basis of weighted levels of the microRNAs forming the outcome signature. RESULTS: Of 305 microRNA probes, 12 (including 5 representing microRNA-181 family members) were associated with event-free survival in the training group (P<0.005). In the validation group, the microRNA summary value was inversely associated with event-free survival (P=0.03). In multivariable analysis, the microRNA summary value remained associated with event-free survival (P=0.04) after adjustment for the allelic ratio of FLT3-ITD to wild-type FLT3 and for the white-cell count. Using results of gene-expression microarray analysis, we found that expression levels of the microRNA-181 family were inversely correlated with expression levels of predicted target genes encoding proteins involved in pathways of innate immunity mediated by toll-like receptors and interleukin-1beta. CONCLUSIONS: A microRNA signature in molecularly defined, high-risk, cytogenetically normal AML is associated with the clinical outcome and with target genes encoding proteins involved in specific innate-immunity pathways.


Asunto(s)
Expresión Génica , Leucemia Mieloide Aguda/genética , MicroARNs/metabolismo , ARN Neoplásico/metabolismo , Adulto , Análisis de Varianza , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica/genética , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Humanos , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/patología , Persona de Mediana Edad , Mutación , Proteínas Nucleares/genética , Nucleofosmina , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Modelos de Riesgos Proporcionales , Sondas ARN , Transactivadores/genética , Transactivadores/metabolismo , Regulador Transcripcional ERG , Tirosina Quinasa 3 Similar a fms/genética
2.
Blood ; 111(11): 5371-9, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18378853

RESUMEN

BAALC expression is considered an independent prognostic factor in cytogenetically normal acute myeloid leukemia (CN-AML), but has yet to be investigated together with multiple other established prognostic molecular markers in CN-AML. We analyzed BAALC expression in 172 primary CN-AML patients younger than 60 years of age, treated similarly on CALGB protocols. High BAALC expression was associated with FLT3-ITD (P = .04), wild-type NPM1 (P < .001), mutated CEBPA (P = .003), MLL-PTD (P = .009), absent FLT3-TKD (P = .005), and high ERG expression (P = .05). In multivariable analysis, high BAALC expression independently predicted lower complete remission rates (P = .04) when adjusting for ERG expression and age, and shorter survival (P = .04) when adjusting for FLT3-ITD, NPM1, CEBPA, and white blood cell count. A gene-expression signature of 312 probe sets differentiating high from low BAALC expressers was identified. High BAALC expression was associated with overexpression of genes involved in drug resistance (MDR1) and stem cell markers (CD133, CD34, KIT). Global microRNA-expression analysis did not reveal significant differences between BAALC expression groups. However, an analysis of microRNAs that putatively target BAALC revealed a potentially interesting inverse association between expression of miR-148a and BAALC. We conclude that high BAALC expression is an independent adverse prognostic factor and is associated with a specific gene-expression profile.


Asunto(s)
Biomarcadores de Tumor/análisis , Expresión Génica , Leucemia Mieloide Aguda/metabolismo , Proteínas de Neoplasias/biosíntesis , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Perfilación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/mortalidad , Nucleofosmina , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Resultado del Tratamiento
3.
Clin Cancer Res ; 14(12): 3889-95, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559610

RESUMEN

PURPOSE: Inhibition of ribonucleotide reductase reduces the availability of the endogenous pool of deoxycytidine and may increase cytarabine (AraC) cytotoxicity. We performed a phase I dose escalation trial of AraC combined with GTI-2040, a 20-mer antisense oligonucleotide shown in preclinical studies to decrease levels of the R2 subunit of ribonucleotide reductase, to determine the maximum tolerated dose in adults with relapsed/refractory acute myeloid leukemia. EXPERIMENTAL DESIGN: Twenty-three adults (ages 18-59 years) were enrolled in this dose escalation phase I trial, receiving high-dose AraC twice daily combined with infusional GTI-2040. An ELISA-based assay measured plasma and intracellular concentrations of GTI-2040. R2 protein changes were evaluated by immunoblotting in pretreatment and post-treatment bone marrow samples. RESULTS: The maximum tolerated dose was 5 mg/kg/d GTI-2040 (days 1-6) and 3 g/m2/dose AraC every 12 hours for 8 doses. Neurotoxicity was dose limiting. Eight patients (35%) achieved complete remission. Mean bone marrow intracellular concentration of GTI-2040 were higher at 120 hours than at 24 hours from the start of GTI-2040 (P = 0.002), suggesting intracellular drug accumulation over time. Reductions in bone marrow levels of R2 protein (>50%) were observed at 24 and 120 hours. Higher baseline R2 protein expression (P = 0.03) and reductions after 24 hours of GTI-2040 (P = 0.04) were associated with complete remission. CONCLUSIONS: GTI-2040 and high-dose AraC were coadministered safely with successful reduction of the intended R2 target and encouraging clinical results. The clinical efficacy of this combination will be tested in an upcoming phase II study.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Citarabina/administración & dosificación , Leucemia Mieloide Aguda/tratamiento farmacológico , Oligodesoxirribonucleótidos/administración & dosificación , Ribonucleótido Reductasas/antagonistas & inhibidores , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Citarabina/efectos adversos , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Oligodesoxirribonucleótidos/efectos adversos , Oligodesoxirribonucleótidos/farmacocinética , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/efectos adversos , Resultado del Tratamiento
4.
Cancer Res ; 65(4): 1277-84, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15735013

RESUMEN

The translocation t(8;21)(q22;q22) in acute myeloid leukemia (AML) results in the expression of the fusion protein RUNX1/MTG8, which in turn recruits histone deacetylases (HDAC) to silence RUNX1 target genes [e.g., interleukin-3 (IL-3)]. We previously reported that expression of the RUNX1/MTG8 target gene IL-3 is synergistically restored by the combination of inhibitors of HDACs (i.e., depsipeptide) and DNA methyltransferases (DNMT; i.e., decitabine) in RUNX1/MTG8-positive Kasumi-1 cells. Thus, we hypothesized that DNMT1 is also part of the transcriptional repressor complex recruited by RUNX1/MTG8. By a chromatin immunoprecipitation assay, we identified a RUNX1/MTG8-DNMT1 complex on the IL-3 promoter in Kasumi-1 cells and in primary RUNX1/MTG8-positive AML blasts. The physical association of RUNX1/MTG8 with DNMT1 was shown by coimmunoprecipitation experiments. Furthermore, RUNX1/MTG8 and DNMT1 were concurrently released from the IL-3 promoter by exposure to depsipeptide or stabilized on the promoter by decitabine treatment. Finally, we proved that RUNX1/MTG8 and DNMT1 were functionally interrelated by showing an enhanced repression of IL-3 after coexpression in 293T cells. These results suggest a novel mechanism for gene silencing mediated by RUNX1/MTG8 and support the combination of HDAC and DNMT inhibitors as a novel therapeutic approach for t(8;21) AML.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Proteínas de Unión al ADN/fisiología , Leucemia Mieloide/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Factores de Transcripción/fisiología , Enfermedad Aguda , Línea Celular , Línea Celular Tumoral , Subunidad alfa 2 del Factor de Unión al Sitio Principal , ADN (Citosina-5-)-Metiltransferasa 1 , Metilación de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Silenciador del Gen , Humanos , Interleucina-3/genética , Leucemia Mieloide/enzimología , Leucemia Mieloide/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteína 1 Compañera de Translocación de RUNX1 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética/fisiología , Transfección
5.
J Clin Oncol ; 26(31): 5078-87, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18809607

RESUMEN

PURPOSE: To evaluate the prognostic significance of CEBPA mutations in the context of established molecular markers in cytogenetically normal (CN) acute myeloid leukemia (AML) and gain biologic insights into leukemogenesis of the CN-AML molecular high-risk subset (FLT3 internal tandem duplication [ITD] positive and/or NPM1 wild type) that has a significantly higher incidence of CEBPA mutations than the molecular low-risk subset (FLT3-ITD negative and NPM1 mutated). PATIENTS AND METHODS: One hundred seventy-five adults age less than 60 years with untreated primary CN-AML were screened before treatment for CEBPA, FLT3, MLL, WT1, and NPM1 mutations and BAALC and ERG expression levels. Gene and microRNA (miRNA) expression profiles were obtained for the CN-AML molecular high-risk patients. RESULTS: CEBPA mutations predicted better event-free (P = .007), disease-free (P = .014), and overall survival (P < .001) independently of other molecular and clinical prognosticators. Among patients with CEBPA mutations, 91% were in the CN-AML molecular high-risk group. Within this group, CEBPA mutations predicted better event-free (P < .001), disease-free (P = .004), and overall survival (P = .009) independently of other molecular and clinical characteristics and were associated with unique gene and miRNA expression profiles. The major features of these profiles were upregulation of genes (eg, GATA1, ZFPM1, EPOR, and GFI1B) and miRNAs (ie, the miR-181 family) involved in erythroid differentiation and downregulation of homeobox genes. CONCLUSION: Pretreatment testing for CEBPA mutations identifies CN-AML patients with different outcomes, particularly in the molecular high-risk group, thus improving molecular risk-based classification of this large cytogenetic subset of AML. The gene and miRNA expression profiling provided insights into leukemogenesis of the CN-AML molecular high-risk group, indicating that CEBPA mutations are associated with partial erythroid differentiation.


Asunto(s)
Biomarcadores de Tumor/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/genética , MicroARNs/análisis , Mutación , Adolescente , Adulto , Supervivencia sin Enfermedad , Femenino , Perfilación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina , Humanos , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/terapia , Masculino , Persona de Mediana Edad , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Nucleofosmina , Medición de Riesgo , Factores de Tiempo , Transactivadores/genética , Regulador Transcripcional ERG , Resultado del Tratamiento , Proteínas WT1/genética , Tirosina Quinasa 3 Similar a fms/genética
6.
Blood ; 111(4): 2364-73, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18083845

RESUMEN

Bortezomib reversibly inhibits 26S proteasomal degradation, interferes with NF-kappaB, and exhibits antitumor activity in human malignancies. Zinc finger protein Sp1 transactivates DNMT1 gene in mice and is functionally regulated through protein abundance, posttranslational modifications (ie, ubiquitination), or interaction with other transcription factors (ie, NF-kappaB). We hypothesize that inhibition of proteasomal degradation and Sp1/NF-kappaB-mediated transactivation may impair aberrant DNA methyltransferase activity. We show here that, in addition to inducing accumulation of polyubiquitinated proteins and abolishment of NF-kappaB activities, bortezomib decreases Sp1 protein levels, disrupts the physical interaction of Sp1/NF-kappaB, and prevents binding of the Sp1/NF-kappaB complex to the DNMT1 gene promoter. Abrogation of Sp1/NF-kappaB complex by bortezomib causes transcriptional repression of DNMT1 gene and down-regulation of DNMT1 protein, which in turn induces global DNA hypomethylation in vitro and in vivo and re-expression of epigenetically silenced genes in human cancer cells. The involvement of Sp1/NF-kappaB in DNMT1 regulation is further demonstrated by the observation that Sp1 knockdown using mithramycin A or shRNA decreases DNMT1 protein levels, which instead are increased by Sp1 or NF-kappaB overexpression. Our results unveil the Sp1/NF-kappaB pathway as a modulator of DNA methyltransferase activity in human cancer and identify bortezomib as a novel epigenetic-targeting drug.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Leucemia Mieloide Aguda/genética , FN-kappa B/fisiología , Pirazinas/farmacología , Transcripción Genética/efectos de los fármacos , Bortezomib , ADN (Citosina-5-)-Metiltransferasas/efectos de los fármacos , Humanos , Proteínas Quinasas/efectos de los fármacos , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo
7.
J Pharmacol Exp Ther ; 321(3): 953-60, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17389244

RESUMEN

In t(8;21) acute myeloid leukemia (AML), the AML1/ETO fusion protein promotes leukemogenesis by recruiting class I histone deacetylase (HDAC)-containing repressor complex to the promoter of AML1 target genes. Valproic acid (VPA), a commonly used antiseizure and mood stabilizer drug, has been shown to cause growth arrest and induce differentiation of malignant cells via HDAC inhibition. VPA causes selective proteasomal degradation of HDAC2 but not other class I HDACs (i.e., HDAC 1, 3, and 8). Therefore, we raised the question of whether this drug can effectively target the leukemogenic activity of the AML1/ETO fusion protein that also recruits HDAC1, a key regulator of normal and aberrant histone acetylation. We report here that VPA treatment disrupts the AML1/ETO-HDAC1 physical interaction, stimulates the global dissociation of AML1/ETO-HDAC1 complex from the promoter of AML1/ETO target genes, and induces relocation of both AML1/ETO and HDAC1 protein from nuclear to perinuclear region. Furthermore, we show that mechanistically these effects associate with a significant inhibition of HDAC activity, histone H3 and H4 hyperacetylation, and recruitment of RNA polymerase II, leading to transcriptional reactivation of target genes (i.e., IL-3) otherwise silenced by AML1/ETO fusion protein. Ultimately, these pharmacological effects resulted in significant antileukemic activity mediated by partial cell differentiation and caspase-dependent apoptosis. Taken together, these data support the notion that VPA might effectively target AML1/ETO-driven leukemogenesis through disruption of aberrant HDAC1 function and that VPA should be integrated in novel therapeutic approaches for AML1/ETO-positive AML.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Histona Desacetilasas/metabolismo , Proteínas de Fusión Oncogénica/genética , Ácido Valproico/farmacología , Acetilación/efectos de los fármacos , Clorometilcetonas de Aminoácidos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Inmunoprecipitación de Cromatina , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , ADN/metabolismo , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Histona Desacetilasa 1 , Histona Desacetilasa 2 , Inhibidores de Histona Desacetilasas , Histonas/metabolismo , Humanos , Interleucina-3/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Proteínas de Fusión Oncogénica/metabolismo , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica/efectos de los fármacos , ARN Polimerasa II/metabolismo , Proteína 1 Compañera de Translocación de RUNX1 , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo
8.
J Clin Oncol ; 25(22): 3337-43, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17577018

RESUMEN

PURPOSE: To validate ERG overexpression as an adverse predictor and assess its prognostic value in the context of other molecular markers in cytogenetically normal (CN) -acute myeloid leukemia (AML). PATIENTS AND METHODS: Seventy-six adult patients with primary CN-AML, younger than 60 years and treated on Cancer and Leukemia Group B (CALGB) trial 19808, were evaluated for ERG expression by quantitative reverse transcriptase polymerase chain reaction. They were then combined with 72 patients enrolled onto CALGB 9621 for analyses that included other molecular markers. RESULTS: Similar to patients enrolled onto CALGB 9621, high ERG expressers on CALGB 19808 had fewer complete remissions (CRs; P = .03) and worse event-free survival (EFS; P = .016) than low ERG expressers. In the combined set, high expressers (n = 55) had fewer CRs (P = .004) and shorter EFS (P < .001) than low expressers (n = 93). High ERG predicted failure to achieve CR (P = .004) after adjusting for BAALC expression (P = .04) and age (P = .008), and EFS (P = .004) after adjusting for FLT3 internal tandem duplication (ITD; P < .001). Among patients without FLT3-ITD (FLT3-ITD negative), only high ERG predicted shorter EFS (P = .001). Among NPM1-mutated (NPM1 positive) patients, high ERG predicted shorter EFS (P = .003), after adjusting for FLT3-ITD (P < .001). When all three markers were considered together, in the favorable FLT3-ITD-negative/NPM1-positive subset, high ERG was the only factor predicting shorter EFS (P = .002). CONCLUSION: We validated ERG overexpression as an adverse predictor in CN-AML. Moreover, by using ERG expression levels, we improved the previously proposed molecular-risk classification of CN-AML based on the presence or absence of FLT3-ITD and NPM1 mutations, given that we identified subsets with different outcome among FLT3-ITD-negative, NPM1-positive, and FLT3-ITD-negative/NPM1-positive patients.


Asunto(s)
Proteínas de Unión al ADN/genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Leucemia Mieloide Aguda/genética , Transactivadores/genética , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/análisis , Femenino , Humanos , Leucemia Mieloide Aguda/terapia , Masculino , Persona de Mediana Edad , Nucleofosmina , Valor Predictivo de las Pruebas , Pronóstico , Modelos de Riesgos Proporcionales , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Medición de Riesgo , Estadísticas no Paramétricas , Trasplante de Células Madre , Análisis de Supervivencia , Regulador Transcripcional ERG , Regulación hacia Arriba
9.
J Clin Oncol ; 25(25): 3884-91, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17679729

RESUMEN

PURPOSE: To determine an optimal biologic dose (OBD) of decitabine as a single agent and then the maximum-tolerated dose (MTD) of valproic acid (VA) combined with decitabine in acute myeloid leukemia (AML). PATIENTS AND METHODS: Twenty-five patients (median age, 70 years) were enrolled; 12 were untreated and 13 had relapsed AML. To determine an OBD (based on a gene re-expression end point), 14 patients received decitabine alone for 10 days. To determine the MTD, 11 patients received decitabine (at OBD, days 1 through 10) plus dose-escalating VA (days 5 through 21). RESULTS: The OBD of decitabine was 20 mg/m(2)/d intravenously, with limited nonhematologic toxicity. In patients treated with decitabine plus VA, dose-limiting encephalopathy occurred in two of two patients at VA 25 mg/kg/d and one of six patients at VA 20 mg/kg/d. Drug-induced re-expression of estrogen receptor (ER) was associated with clinical response (P < or = .05). ER promoter demethylation, global DNA hypomethylation, depletion of DNA methyltransferase enzyme, and histone hyperacetylation were also observed. In an intent-to-treat analysis, the response rate was 44% (11 of 25). Of 21 assessable patients, 11 (52%) responded: four with morphologic and cytogenetic complete remission (CR; each had complex karyotype), four with incomplete CR, and three with partial remission. In untreated AML, four of nine assessable patients achieved CR. Clinical responses appeared similar for decitabine alone or with VA. CONCLUSION: Low-dose decitabine was safe and showed encouraging clinical and biologic activity in AML, but the addition of VA led to encephalopathy at relatively low doses. On the basis of these results, additional studies of decitabine (20 mg/m(2)/d for 10 days) alone or with an alternative deacetylating agent are warranted.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Azacitidina/análogos & derivados , Encefalopatías/inducido químicamente , Leucemia Mieloide Aguda/tratamiento farmacológico , Ácido Valproico/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Azacitidina/administración & dosificación , Azacitidina/farmacocinética , Decitabina , Fatiga/inducido químicamente , Humanos , Infecciones/inducido químicamente , Dosis Máxima Tolerada , Persona de Mediana Edad , Neutropenia/inducido químicamente , Inducción de Remisión , Insuficiencia del Tratamiento , Ácido Valproico/farmacocinética
10.
Blood ; 109(12): 5164-7, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17341662

RESUMEN

The clinical impact of MLL partial tandem duplication (MLL-PTD) was evaluated in 238 adults aged 18 to 59 years with cytogenetically normal (CN) de novo acute myeloid leukemia (AML) who were treated intensively on similar Cancer and Leukemia Group B protocols 9621 and 19808. Twenty-four (10.1%) patients harbored an MLL-PTD. Of those, 92% achieved complete remission (CR) compared with 83% of patients without MLL-PTD (P=.39). Neither overall survival nor disease-free survival significantly differed between the 2 groups (P=.67 and P=.55, respectively). Thirteen MLL-PTD(+) patients relapsed within 1.4 years of achieving CR. MLL-PTD(+) patients who relapsed more often had other adverse CN-AML-associated molecular markers. In contrast with previously reported studies, 9 (41%) MLL-PTD(+) patients continue in long-term first remission (CR1; range, 2.5-7.7 years). Intensive consolidation therapy that included autologous peripheral stem-cell transplantation during CR1 may have contributed to the better outcome of this historically poor-prognosis group of CN-AML patients with MLL-PTD.


Asunto(s)
Leucemia Mieloide/genética , Leucemia Mieloide/mortalidad , Proteína de la Leucemia Mieloide-Linfoide/genética , Secuencias Repetidas en Tándem , Enfermedad Aguda , Adolescente , Adulto , Análisis Citogenético , Supervivencia sin Enfermedad , N-Metiltransferasa de Histona-Lisina , Humanos , Persona de Mediana Edad , Trasplante de Células Madre de Sangre Periférica , Inducción de Remisión , Análisis de Supervivencia , Sobrevivientes
11.
J Clin Oncol ; 24(24): 3904-11, 2006 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-16921041

RESUMEN

PURPOSE: To analyze the prognostic impact of mutated KIT (mutKIT) in core-binding factor acute myeloid leukemia (AML) with inv(16)(p13q22) and t(8;21)(q22;q22). PATIENTS AND METHODS: Sixty-one adults with inv(16) and 49 adults with t(8;21), assigned to postremission therapy with repetitive cycles of higher dose cytarabine were analyzed for mutKIT in exon 17 (mutKIT17) and 8 (mutKIT8) by denaturing high-performance liquid chromatography and direct sequencing at diagnosis. The median follow-up was 5.3 years. RESULTS: Among patients with inv(16), 29.5% had mutKIT (16% with mutKIT17 and 13% with sole mutKIT8). Among patients with t(8;21), 22% had mutKIT (18% with mutKIT17 and 4% with sole mutKIT8). Complete remission rates of patients with mutKIT and wild-type KIT (wtKIT) were similar in both cytogenetic groups. In inv(16), the cumulative incidence of relapse (CIR) was higher for patients with mutKIT (P = .05; 5-year CIR, 56% v 29%) and those with mutKIT17 (P = .002; 5-year CIR, 80% v 29%) compared with wtKIT patients. Once data were adjusted for sex, mutKIT predicted worse overall survival (OS). In t(8;21), mutKIT predicted higher CIR (P = .017; 5-year CIR, 70% v 36%), but did not influence OS. CONCLUSION: We report for the first time that mutKIT, and particularly mutKIT17, confer higher relapse risk, and both mutKIT17 and mutKIT8 appear to adversely affect OS in AML with inv(16). We also confirm the adverse impact of mutKIT on relapse risk in t(8;21) AML. We suggest that patients with core-binding factor AML should be screened for mutKIT at diagnosis for both prognostic and therapeutic purposes, given that activated KIT potentially can be targeted with novel tyrosine kinase inhibitors.


Asunto(s)
Inversión Cromosómica , Leucemia Mieloide/genética , Mutación , Proteínas Proto-Oncogénicas c-kit/genética , Translocación Genética , Enfermedad Aguda , Adulto , Cromatografía Líquida de Alta Presión , Femenino , Humanos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Valor Predictivo de las Pruebas , Pronóstico , Análisis de Supervivencia
12.
Blood ; 108(5): 1677-83, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16670265

RESUMEN

Patients with acute myeloid leukemia (AML) and normal karyotype are classified in an intermediate-risk group, albeit this subset is heterogeneous for clinical outcome. A recent complementary DNA microarray study identified a gene-expression signature that--when used to cluster normal karyotype patients--separated them into 2 prognostically relevant subgroups. We sought the first independent validation of the prognostic value of this signature. Using oligonucleotide microarrays to measure gene expression in samples from uniformly treated adults with karyotypically normal AML, we performed cluster analysis based on the previously identified signature. We also developed a well-defined classification rule using the signature to predict outcome for individual patients. Cluster analysis confirmed the prognostic utility of the signature: patient clusters differed in overall (P = .001) and disease-free (P = .001) survival. The signature-based classifier identified groups with differences in overall (P = .02) and disease-free (P = .05) survival. A strong association of the outcome classifier with the prognostically adverse FLT3 internal tandem duplication (FLT3 ITD) potentially explained the prognostic significance of the signature. However, in the subgroup of patients without FLT3 ITD there was a moderate difference in survival for the classifier-derived groups. Our analysis confirms the applicability of the gene-expression profiling strategy for outcome prediction in cytogenetically normal AML.


Asunto(s)
Leucemia Mieloide/genética , Leucemia Mieloide/terapia , Enfermedad Aguda , Adulto , Algoritmos , Análisis por Conglomerados , Etnicidad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Cariotipificación , Leucemia Mieloide/mortalidad , Masculino , Persona de Mediana Edad , Pronóstico , Análisis de Supervivencia , Resultado del Tratamiento
13.
Blood ; 106(10): 3380-2, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16076875

RESUMEN

Adult T-cell leukemia (ATL) is a highly chemoresistant and usually fatal T-cell malignancy due to the human T-cell lymphotropic virus-1 (HTLV-1). After chemotherapy failure, antiretrovirals and interferon-alpha (IFN-alpha) produce brief responses followed by progression and death. More effective agents and new approaches to detect and treat minimal residual disease are needed. ATL cells express CD52, the target of the antibody alemtuzumab, which is active in a preclinical model of ATL and is cytotoxic for p53-deficient cells. A patient with refractory chronic ATL in transformation achieved longer than a 1-year complete hematologic response following 12 weeks of outpatient subcutaneous alemtuzumab. Persistent suppression of HTLV-1 viral load, even at recovery of T cells, after alemtuzumab and efficient in vitro complement-mediated cytotoxicity of primary ATL cells with mutated TP53 were observed. The unprecedented response and the profound suppression of HTLV-1 viral load observed in this patient suggest that further clinical investigation of alemtuzumab in ATL is warranted.


Asunto(s)
Fármacos Anti-VIH/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Antineoplásicos/administración & dosificación , Antineoplásicos/administración & dosificación , Virus Linfotrópico T Tipo 1 Humano , Interferón-alfa/administración & dosificación , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Zidovudina/administración & dosificación , Alemtuzumab , Anticuerpos Monoclonales Humanizados , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Antígeno CD52 , Femenino , Glicoproteínas/metabolismo , Humanos , Leucemia-Linfoma de Células T del Adulto/metabolismo , Leucemia-Linfoma de Células T del Adulto/virología , Persona de Mediana Edad , Recuperación de la Función/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T/virología , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/metabolismo , Carga Viral/métodos
14.
Blood ; 106(1): 345-52, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15774615

RESUMEN

MLL (ALL-1) chimeric fusions and MLL partial tandem duplications (PTD) may have mechanistically distinct contributions to leukemogenesis. Acute myeloid leukemia (AML) blasts with the t(9;11)(p22; q23) express MLL-AF9 and MLL wild-type (WT) transcripts, while normal karyotype AML blasts with the MLL(PTD/WT) genotype express MLL PTD but not the MLL WT. Silencing of MLL WT in MLL(PTD/WT) blasts was reversed by DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors, and MLL WT induction was associated with selective sensitivity to cell death. Reduction of MLL PTD expression induced MLL WT and reduced blast colony-forming units, supporting opposing functions for MLL PTD and MLL WT whereby the MLL PTD contributes to the leukemic phenotype via a recessive gain-of-function. The coincident suppression of the MLL WT allele with the expression of the MLL PTD allele, along with the functional data presented here, supports the hypothesis that loss of WT MLL function via monoallelic repression contributes to the leukemic phenotype by the remaining mutant allele. These data from primary AML and the pharmacologic reversal of MLL WT silencing associated with a favorable alteration in the threshold for apoptosis suggest that these patients with poor prognosis may benefit from demethylating or histone deacetylase inhibitor therapy, or both.


Asunto(s)
Metilasas de Modificación del ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Duplicación de Gen , Inhibidores de Histona Desacetilasas , Leucemia Mieloide/genética , Proto-Oncogenes/genética , Factores de Transcripción/genética , Enfermedad Aguda , Muerte Celular , Islas de CpG/genética , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Regulación Leucémica de la Expresión Génica , Genotipo , N-Metiltransferasa de Histona-Lisina , Humanos , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/patología , Proteína de la Leucemia Mieloide-Linfoide , Oligodesoxirribonucleótidos , Fenotipo , Secuencias Repetidas en Tándem , Células Tumorales Cultivadas
15.
Am J Pathol ; 163(6): 2277-87, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14633602

RESUMEN

To determine the role of beta-catenin pathway in human skin carcinogenesis, 135 non-melanoma skin tumors were analyzed for beta-catenin expression and gene mutations. Intense nucleo-cytoplasmic immunoreactivity for C terminus beta-catenin antibodies was observed in all pilomatricomas and in single cases of trichoepithelioma and squamous cell carcinoma showing peculiar signs of matrical differentiation. Moderate increase of beta-catenin nuclear staining was detected in a significant proportion of basal cell carcinomas, Bowen disease, spiroadenomas, and occasionally also in squamous cell carcinomas, but in these neoplasms only a limited fraction of tumor cells accumulated beta-catenin. Molecular analysis revealed that beta-catenin gene mutations are a peculiar feature of skin tumors with matrical differentiation and correlate with a pattern of intense and diffuse beta-catenin nuclear expression. In contrast, adenomatous polyposis coli (APC) and AXIN2 mutations were not involved in skin tumorigenesis. Analysis of Wnt pathway revealed that TCF-1 and MITF-M were selectively induced in the tumor types harboring beta-catenin mutations, indicating that a Wnt/beta-catenin pathway involving TCF-1 and MITF-M is activated in these tumors. Interestingly, high expression levels of TCF-3 were found in basal cell carcinomas and spiroadenomas. TCF-3 is reported to act as a negative modulator of beta-catenin degradation pathway. Thus, the moderate increase of beta-catenin nuclear staining detected in these tumor types might, at least in part, be due to a TCF-3-dependent mechanism. Finally, we found that the presence of beta-catenin mutations significantly correlated with loss of nuclear immunoreactivity for an antibody raised against the N terminus of beta-catenin (alphaABC). Thus, a combined analysis with C terminus-beta-catenin antibodies and alphaABC Ab may represent a powerful investigative approach for the detection of beta-catenin structural alterations.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Mutación , Neoplasias Cutáneas/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Anticuerpos Monoclonales , Proteína Axina , Proteínas del Citoesqueleto/inmunología , Análisis Mutacional de ADN , Genes APC , Humanos , Inmunohistoquímica , Piel/metabolismo , Transactivadores/inmunología , beta Catenina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA