Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 128(7): 902-10, 2016 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-27335276

RESUMEN

In myelodysplastic syndromes (MDSs), the evolution of risk for disease progression or death has not been systematically investigated despite being crucial for correct interpretation of prognostic risk scores. In a multicenter retrospective study, we described changes in risk over time, the consequences for basal prognostic scores, and their potential clinical implications. Major MDS prognostic risk scoring systems and their constituent individual predictors were analyzed in 7212 primary untreated MDS patients from the International Working Group for Prognosis in MDS database. Changes in risk of mortality and of leukemic transformation over time from diagnosis were described. Hazards regarding mortality and acute myeloid leukemia transformation diminished over time from diagnosis in higher-risk MDS patients, whereas they remained stable in lower-risk patients. After approximately 3.5 years, hazards in the separate risk groups became similar and were essentially equivalent after 5 years. This fact led to loss of prognostic power of different scoring systems considered, which was more pronounced for survival. Inclusion of age resulted in increased initial prognostic power for survival and less attenuation in hazards. If needed for practicability in clinical management, the differing development of risks suggested a reasonable division into lower- and higher-risk MDS based on the IPSS-R at a cutoff of 3.5 points. Our data regarding time-dependent performance of prognostic scores reflect the disparate change of risks in MDS subpopulations. Lower-risk patients at diagnosis remain lower risk whereas initially high-risk patients demonstrate decreasing risk over time. This change of risk should be considered in clinical decision making.


Asunto(s)
Transformación Celular Neoplásica/patología , Síndromes Mielodisplásicos/mortalidad , Anciano , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Factores de Riesgo , Factores de Tiempo , Organización Mundial de la Salud
2.
Biol Blood Marrow Transplant ; 22(7): 1212-1217, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27044907

RESUMEN

The prognostic relevance of cytogenetics at diagnosis on the outcome of allogeneic hematopoietic stem cell transplantation (alloHCT) for adult acute lymphoblastic leukemia (ALL) remains unclear. We retrospectively analyzed outcomes of 333 adult ALL patients who underwent alloHCT at our institution over a 10-year period. Patients were classified according to disease status at transplantation (complete response [CR] 1 [n = 202] or > CR1) and according to cytogenetic risk, defined as good (2%), intermediate (42%), poor (46%), or unknown (10%) based on available outcome data for each of the cytogenetic abnormalities. Three-year overall survival (OS), leukemia-free survival (LFS), and relapse incidence (RI) were 55.7%, 47.9% and 27.5%, respectively; 1-year nonrelapse mortality (NRM) was 17.3%. For patients undergoing alloHCT in CR1, 3-year OS, LFS, and RI were 69.8%, 62.3%, and 17.1%, respectively. In multivariable analysis, cytogenetic risk did not impact OS or LFS for the whole cohort or for patients who underwent transplantation in CR1. Disease status at alloHCT was an independent predictor for LFS (CR1 versus others: hazard ratio [HR], 3.17; P < .01) and OS (CR1 versus others: HR, 2.90; P < .01). Graft-versus-host disease prophylaxis with tacrolimus/sirolimus was associated with a low NRM of 11.5% in the alloHCT recipients in CR1. Our data indicate that cytogenetic risk is not an independent predictor of outcomes in alloHCT performed to treat adult ALL.


Asunto(s)
Aberraciones Cromosómicas , Trasplante de Células Madre Hematopoyéticas/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Adolescente , Adulto , Anciano , Femenino , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Inmunosupresores/uso terapéutico , Masculino , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Premedicación/métodos , Premedicación/mortalidad , Pronóstico , Estudios Retrospectivos , Medición de Riesgo/métodos , Sirolimus/uso terapéutico , Tacrolimus/uso terapéutico , Trasplante Homólogo , Adulto Joven
3.
Genes Chromosomes Cancer ; 54(12): 809-17, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26355708

RESUMEN

In myelodysplastic syndromes (MDS), deletion of the short arm of chromosome 12 (del(12p)) is usually a small abnormality, rarely detected as a single aberration by chromosome banding analysis (CBA) of bone marrow metaphases. Del(12p) has been described in 0.6 to 5% of MDS patients at initial diagnosis and is associated with a good to intermediate prognosis as a sole anomaly according to current scoring systems. Here, we present the results of a systematic del(12p) testing in a German prospective diagnostic study (clinicaltrials.gov: NCT01355913) on 367 MDS patients in whom CD34+ peripheral blood cells were analysed for the presence of del(12p) by sequential fluorescence in situ hybridization (FISH) analyses. A cohort of 2,902 previously published MDS patients diagnosed by CBA served as control. We demonstrate that, using a sensitive FISH technique, 12p deletion occurs significantly more frequently in MDS than previously described (7.6% by CD34+ PB-FISH vs. 1.6% by CBA, P < 0.001) and is often associated with other aberrations (93% by CD34+ PB-FISH vs. 60% by CBA). Additionally, the detection rate can be increased by repeated analyses in a patient over time which is important for the patient´s prognosis to distinguish a sole anomaly from double or complex aberrations. To our knowledge, this is the first study to screen for 12p deletions with a suitable probe for ETV6/TEL in 12p13. Our data suggest that the supplement of a probe for the detection of a 12p deletion to common FISH probe panels helps to avoid missing a del(12p), especially as part of more complex aberrations.


Asunto(s)
Hibridación Fluorescente in Situ/métodos , Síndromes Mielodisplásicos/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD34/genética , Bandeo Cromosómico , Deleción Cromosómica , Cromosomas Humanos Par 12/genética , Grupos Control , Femenino , Alemania , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Represoras/genética , Adulto Joven , Proteína ETS de Variante de Translocación 6
4.
Haematologica ; 100(2): 205-13, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25344522

RESUMEN

International Prognostic Scoring Systems are used to determine the individual risk profile of myelodysplastic syndrome patients. For the assessment of International Prognostic Scoring Systems, an adequate chromosome banding analysis of the bone marrow is essential. Cytogenetic information is not available for a substantial number of patients (5%-20%) with dry marrow or an insufficient number of metaphase cells. For these patients, a valid risk classification is impossible. In the study presented here, the International Prognostic Scoring Systems were validated based on fluorescence in situ hybridization analyses using extended probe panels applied to cluster of differentiation 34 positive (CD34(+)) peripheral blood cells of 328 MDS patients of our prospective multicenter German diagnostic study and compared to chromosome banding results of 2902 previously published patients with myelodysplastic syndromes. For cytogenetic risk classification by fluorescence in situ hybridization analyses of CD34(+) peripheral blood cells, the groups differed significantly for overall and leukemia-free survival by uni- and multivariate analyses without discrepancies between treated and untreated patients. Including cytogenetic data of fluorescence in situ hybridization analyses of peripheral CD34(+) blood cells (instead of bone marrow banding analysis) into the complete International Prognostic Scoring System assessment, the prognostic risk groups separated significantly for overall and leukemia-free survival. Our data show that a reliable stratification to the risk groups of the International Prognostic Scoring Systems is possible from peripheral blood in patients with missing chromosome banding analysis by using a comprehensive probe panel (clinicaltrials.gov identifier:01355913).


Asunto(s)
Antígenos CD34/sangre , Aberraciones Cromosómicas , Análisis Citogenético/métodos , Hibridación Fluorescente in Situ/métodos , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Humanos , Agencias Internacionales , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/mortalidad , Estadificación de Neoplasias , Pronóstico , Estudios Prospectivos , Tasa de Supervivencia , Adulto Joven
5.
Br J Haematol ; 164(2): 223-32, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24383843

RESUMEN

The standard dose of imatinib for newly diagnosed patients with chronic phase chronic myeloid leukaemia (CP-CML) is 400 mg daily (IM400), but the optimal dose is unknown. This randomized phase II study compared the rates of molecular, haematological and cytogenetic response to IM400 vs. imatinib 400 mg twice daily (IM800) in 153 adult patients with CP-CML. Dose adjustments for toxicity were flexible to maximize retention on study. Molecular response (MR) at 12 months was deeper in the IM800 arm (4-log reduction of BCR-ABL1 mRNA: 25% vs. 10% of patients, P = 0·038; 3-log reduction: 53% vs. 35%, P = 0·049). During the first 12 months BCR-ABL1 levels in the IM800 arm were an average 2·9-fold lower than in the IM400 arm (P = 0·010). Complete haematological response was similar, but complete cytogenetic response was higher with IM800 (85% vs. 67%, P = 0·040). Grade 3-4 toxicities were more common for IM800 (58% vs. 31%, P = 0·0007), and were most commonly haematological. Few patients have relapsed, progressed or died, but both progression-free (P = 0·048) and relapse-free (P = 0·031) survival were superior for IM800. In newly diagnosed CP-CML patients, IM800 induced deeper MRs than IM400, with a trend for improved progression-free and overall survival, but was associated with more severe toxicity.


Asunto(s)
Antineoplásicos/administración & dosificación , Benzamidas/administración & dosificación , Leucemia Mieloide de Fase Crónica/tratamiento farmacológico , Piperazinas/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirimidinas/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Benzamidas/efectos adversos , Femenino , Proteínas de Fusión bcr-abl/genética , Humanos , Mesilato de Imatinib , Quimioterapia de Inducción , Leucemia Mieloide de Fase Crónica/genética , Leucemia Mieloide de Fase Crónica/mortalidad , Masculino , Persona de Mediana Edad , Mutación , Piperazinas/efectos adversos , Dominios y Motivos de Interacción de Proteínas/genética , Inhibidores de Proteínas Quinasas/efectos adversos , Pirimidinas/efectos adversos , Resultado del Tratamiento , Adulto Joven
6.
Blood ; 120(12): 2454-65, 2012 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-22740453

RESUMEN

The International Prognostic Scoring System (IPSS) is an important standard for assessing prognosis of primary untreated adult patients with myelodysplastic syndromes (MDS). To refine the IPSS, MDS patient databases from international institutions were coalesced to assemble a much larger combined database (Revised-IPSS [IPSS-R], n = 7012, IPSS, n = 816) for analysis. Multiple statistically weighted clinical features were used to generate a prognostic categorization model. Bone marrow cytogenetics, marrow blast percentage, and cytopenias remained the basis of the new system. Novel components of the current analysis included: 5 rather than 3 cytogenetic prognostic subgroups with specific and new classifications of a number of less common cytogenetic subsets, splitting the low marrow blast percentage value, and depth of cytopenias. This model defined 5 rather than the 4 major prognostic categories that are present in the IPSS. Patient age, performance status, serum ferritin, and lactate dehydrogenase were significant additive features for survival but not for acute myeloid leukemia transformation. This system comprehensively integrated the numerous known clinical features into a method analyzing MDS patient prognosis more precisely than the initial IPSS. As such, this IPSS-R should prove beneficial for predicting the clinical outcomes of untreated MDS patients and aiding design and analysis of clinical trials in this disease.


Asunto(s)
Médula Ósea/patología , Análisis Citogenético , Síndromes Mielodisplásicos/clasificación , Síndromes Mielodisplásicos/mortalidad , Pancitopenia/diagnóstico , Pancitopenia/mortalidad , Adulto , Anciano , Anciano de 80 o más Años , Aberraciones Cromosómicas , Femenino , Humanos , Agencias Internacionales , Cariotipificación , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/patología , Pronóstico , Medición de Riesgo , Factores de Riesgo , Tasa de Supervivencia
7.
Genet Med ; 15(6): 484-94, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23619274

RESUMEN

Microarray methodologies, to include array comparative genomic hybridization and single-nucleotide polymorphism-based arrays, are innovative methods that provide genomic data. These data should be correlated with the results from the standard methods, chromosome and/or fluorescence in situ hybridization, to ascertain and characterize the genomic aberrations of neoplastic disorders, both liquid and solid tumors. Over the past several decades, standard methods have led to an accumulation of genetic information specific to many neoplasms. This specificity is now used for the diagnosis and classification of neoplasms. Cooperative studies have revealed numerous correlations between particular genetic aberrations and therapeutic outcomes. Molecular investigation of chromosomal abnormalities identified by standard methods has led to discovery of genes, and gene function and dysfunction. This knowledge has led to improved therapeutics and, in some disorders, targeted therapies. Data gained from the higher-resolution microarray methodologies will enhance our knowledge of the genomics of specific disorders, leading to more effective therapeutic strategies. To assist clinical laboratories in validation of the methods, their consistent use, and interpretation and reporting of results from these microarray methodologies, the American College of Medical Genetics and Genomics has developed the following professional standard and guidelines.


Asunto(s)
Aberraciones Cromosómicas , Neoplasias/diagnóstico , Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/normas , Genómica/instrumentación , Genómica/métodos , Genómica/normas , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos/instrumentación , Reproducibilidad de los Resultados , Programas Informáticos
8.
Blood ; 117(22): 5918-30, 2011 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-21471525

RESUMEN

We studied leukemic stem cells (LSCs) in a Smad4(-/-) mouse model of acute myelogenous leukemia (AML) induced either by the HOXA9 gene or by the fusion oncogene NUP98-HOXA9. Although Hoxa9-Smad4 complexes accumulate in the cytoplasm of normal hematopoietic stem cells and progenitor cells (HSPCs) transduced with these oncogenes, there is no cytoplasmic stabilization of HOXA9 in Smad4(-/-) HSPCs, and as a consequence increased levels of Hoxa9 is observed in the nucleus leading to increased immortalization in vitro. Loss of Smad4 accelerates the development of leukemia in vivo because of an increase in transformation of HSPCs. Therefore, the cytoplasmic binding of Hoxa9 by Smad4 is a mechanism to protect Hoxa9-induced transformation of normal HSPCs. Because Smad4 is a potent tumor suppressor involved in growth control, we developed a strategy to modify the subcellular distribution of Smad4. We successfully disrupted the interaction between Hoxa9 and Smad4 to activate the TGF-ß pathway and apoptosis, leading to a loss of LSCs. Together, these findings reveal a major role for Smad4 in the negative regulation of leukemia initiation and maintenance induced by HOXA9/NUP98-HOXA9 and provide strong evidence that antagonizing Smad4 stabilization by these oncoproteins might be a promising novel therapeutic approach in leukemia.


Asunto(s)
Núcleo Celular/metabolismo , Transformación Celular Neoplásica , Citoplasma/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/fisiología , Proteína Smad4/fisiología , Animales , Apoptosis , Western Blotting , Trasplante de Médula Ósea , Inmunoprecipitación de Cromatina , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/genética , Humanos , Inmunoprecipitación , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
10.
Hum Mol Genet ; 19(8): 1507-14, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20093295

RESUMEN

Myelodysplastic syndromes and acute myeloid leukemia with an isodicentric X chromosome [idic(X)(q13)] occur in elderly women and frequently display ringed sideroblasts. Because of the rarity of idic(X)(q13), little is known about its formation, whether a fusion gene is generated, and patterns of additional aberrations. We here present an SNP array study of 14 idic(X)-positive myeloid malignancies, collected through an international collaborative effort. The breakpoints clustered in two regions of segmental duplications and were not in a gene, making dosage effects from the concurrent gain of Xpter-q13 and loss of Xq13-qter, rather than a fusion gene, the most likely pathogenetic outcome. Methylation analysis revealed involvement of the inactive X chromosomes in five cases and of the active in two. The ABCB7 gene, mutated in X-linked sideroblastic anemia and spinocerebellar ataxia, is in the deleted region, suggesting that loss of this gene underlies the frequent presence of ringed sideroblasts. Additional genetic abnormalities were present in 12/14 (86%), including partial uniparental disomies for 9p (one case) and 4q (two cases) associated with homozygous mutations of JAK2 and TET2, respectively. In total, TET2 mutations were seen in 4/11 (36%) analyzed cases, thus constituting a common secondary event in idic(X)-positive malignancies.


Asunto(s)
Cromosomas Humanos X/genética , Roturas del ADN , Proteínas de Unión al ADN/genética , Mutación , Síndromes Mielodisplásicos/genética , Proteínas Proto-Oncogénicas/genética , Duplicaciones Segmentarias en el Genoma , Anciano , Anciano de 80 o más Años , Cromosomas Humanos X/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Femenino , Humanos , Persona de Mediana Edad , Síndromes Mielodisplásicos/metabolismo , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas/metabolismo
11.
Blood ; 115(12): 2462-72, 2010 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-20075157

RESUMEN

The GFI1 gene encodes a transcriptional repressor, which regulates myeloid differentiation. In the mouse, Gfi1 deficiency causes neutropenia and an accumulation of granulomonocytic precursor cells that is reminiscent of a myelodysplastic syndrome. We report here that a variant allele of GFI1 (GFI1(36N)) is associated with acute myeloid leukemia (AML) in white subjects with an odds ratio of 1.6 (P < 8 x 10(-5)). The GFI1(36N) variant occurred in 1806 AML patients with an allele frequency of 0.055 compared with 0.035 in 1691 healthy control patients in 2 independent cohorts. We observed that both GFI1 variants maintain the same activity as transcriptional repressors but differ in their regulation by the AML1/ETO (RUNX1/RUNX1T1) fusion protein produced in AML patients with a t(8;21) translocation. AML1/ETO interacts and colocalizes with the more common GFI1(36S) form in the nucleus and inhibits its repressor activity. However, the variant GFI1(36N) protein has a different subnuclear localization than GFI1(36S). As a consequence, AML1/ETO does not colocalize with GFI1(36N) and is unable to inhibit its repressor activity. We conclude that both variants of GFI1 differ in their ability to be regulated by interacting proteins and that the GFI1(36N) variant form exhibits distinct biochemical features that may confer a predisposition to AML.


Asunto(s)
Proteínas de Unión al ADN/genética , Predisposición Genética a la Enfermedad , Leucemia Mieloide Aguda/genética , Polimorfismo de Nucleótido Simple , Factores de Transcripción/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Células COS , Núcleo Celular/metabolismo , Chlorocebus aethiops , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Frecuencia de los Genes , Variación Genética , Células HeLa , Humanos , Leucemia Mieloide Aguda/metabolismo , Desequilibrio de Ligamiento , Masculino , Ratones , Persona de Mediana Edad , Células 3T3 NIH , Proteínas de Fusión Oncogénica/metabolismo , Proteína 1 Compañera de Translocación de RUNX1 , Factores de Transcripción/metabolismo , Translocación Genética , Adulto Joven
12.
Br J Haematol ; 148(1): 48-58, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19821823

RESUMEN

Attempts to overcome multi-drug resistance in acute myeloid leukaemia (AML) have been limited by toxicities. To investigate the effect of reducing peak drug levels, we performed sequential phase II studies using continuous infusion daunorubicin and cytarabine without (AD) and then with ciclosporin (ADC) in older patients with AML. Untreated patients (age 56+ years) received daunorubicin (45 mg/m2 per day for 3 d) and cytarabine (200 mg/m2 per day for 7 d), both by continuous infusion, without (S0112, 60 patients) and then with (S0301, 50 patients) the addition of ciclosporin. Complete response (CR) rates were 38% on S0112 and 44% on S0301. Fatal induction toxicities occurred in 17% and 12% respectively, arising primarily from infection and haemorrhage. Median overall and relapse-free survival was 7 and 8 months for AD respectively, and 6 and 14 months for ADC. Patients with phenotypic or functional P-glycoprotein had somewhat higher CR rates with ADC than AD, although confidence intervals overlapped. In these sequential trials, continuous infusion AD produced CR rates comparable to those with bolus daunorubicin. The addition of ciclosporin did not cause undue toxicities, produced a similar CR rate, and possibly improved relapse-free survival. Further correlate analyses did not identify a subpopulation specifically benefitting from the addition of ciclosporin.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Ciclosporina/administración & dosificación , Ciclosporina/efectos adversos , Citarabina/administración & dosificación , Citarabina/efectos adversos , Daunorrubicina/administración & dosificación , Daunorrubicina/efectos adversos , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Métodos Epidemiológicos , Femenino , Humanos , Infusiones Intravenosas , Leucemia Mieloide Aguda/sangre , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Resultado del Tratamiento
13.
Br J Haematol ; 151(5): 430-4, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21113977

RESUMEN

Clofarabine and cytarabine target different steps in DNA synthesis and replication, are synergistic in vivo, and have non-overlapping toxicities, making this combination a potentially promising treatment for acute lymphocytic leukaemia. Thirty-seven patients were treated. The median age was 41 years, 44% of patients were either in ≥2nd relapse or had refractory disease and 59% of patients had poor risk cytogenetics. Six out of 36 patients (17%) achieved a complete remission with or without complete count recovery; median overall survival was 3 months. Nucleoside transporter expression did not predict outcome. This regimen lacked sufficient activity to warrant further testing.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Nucleótidos de Adenina/administración & dosificación , Nucleótidos de Adenina/efectos adversos , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Arabinonucleósidos/administración & dosificación , Arabinonucleósidos/efectos adversos , Clofarabina , Factor de Crecimiento del Tejido Conjuntivo/sangre , Citarabina/administración & dosificación , Citarabina/efectos adversos , Femenino , Humanos , Masculino , Proteínas de Neoplasias/sangre , Proteínas de Transporte de Nucleósidos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/sangre , Pronóstico , Recurrencia , Resultado del Tratamiento
14.
Breast Cancer Res Treat ; 119(2): 391-8, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19851858

RESUMEN

A serious complication associated with breast cancer treatment is the increased risk for development of therapy-related myeloid neoplasms (t-MN). To determine whether dose-intensive adjuvant regimens for breast cancer induce genetic damage to hematopoietic stem cells, defined by the emergence of clonal hematopoiesis, and whether detection of clonal hematopoiesis could be used as an early marker for the subsequent development of t-MN, the Southwest Oncology Group designed a pilot clonality investigation to estimate the incidence of clonal hematopoiesis during and shortly after completion of the dose intensive neoadjuvant regimens for high-risk breast cancer patients. Peripheral blood samples from 274 patients obtained prior to treatment, at time of surgery, and at 6 and 12 months post-surgery were examined by two different clonality assays: the HUMARA (HUMan Androgen Receptor) assay to estimate the incidence of early genetic damage by clonal proliferation, and microsatellite instability (MSI) testing to screen for LOH or defective DNA mismatch repair mechanisms. Clonal hematopoiesis was negative in 93.5% of the samples analyzed. Five patients showed a HUMARA-positive/MSI-negative pattern, and no patients showed a HUMARA-negative/MSI-positive pattern. With a median follow-up of 3.1 years, one patient in our study developed t-AML at 3 years 5 months after randomization. Our results indicate that clonal hematopoiesis assays performed within the 2 years following dose-intensive neoadjuvant therapy failed to identify an emerging clonal hematopoietic stem cell population. Longer clinical follow-up will be necessary to define better the positive predictive value of detecting clonal hematopoiesis in the HUMARA+/MSI- cases.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias de la Mama/tratamiento farmacológico , Pruebas Genéticas , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Leucemia Mieloide/etiología , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Ensayo de Unidades Formadoras de Colonias , Ciclofosfamida/administración & dosificación , Doxorrubicina/administración & dosificación , Femenino , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Predisposición Genética a la Enfermedad , Hematopoyesis/genética , Células Madre Hematopoyéticas/patología , Humanos , Leucemia Mieloide/inducido químicamente , Leucemia Mieloide/genética , Leucemia Mieloide/patología , Inestabilidad de Microsatélites/efectos de los fármacos , Persona de Mediana Edad , Proyectos Piloto , Valor Predictivo de las Pruebas , Estudios Prospectivos , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo
15.
Blood ; 112(3): 903-9, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18519812

RESUMEN

Allogeneic hematopoietic cell transplantation (HCT) is the only known curative modality for patients with Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph(+) ALL). Sixty-seven patients with HLA-matched sibling donors received fractionated total body irradiation (FTBI) and high-dose VP16, whereas 11 patients received FTBI/VP16/cyclophosphamide, and 1 patient received FTBI/VP16/busulfan. The median age was 36 years. At the time of HCT, 49 patients (62%) were in first complete remission (CR1) and 30 patients (38%) were beyond CR1 (> CR1). The median follow-up was 75 months (range, 14-245 months). The 10-year overall survival for the CR1 and beyond CR1 patients was 54% and 29% (P = .01), respectively, and event-free survival was 48% and 26% (P = .02), respectively. There was no significant difference in relapse incidence (28% vs 41%, P = .28), but nonrelapse mortality was significantly higher in the beyond CR1 patients, (31% vs 54%, P = .03, respectively). By univariate analysis, factors affecting event-free and overall survival were white blood cell count at diagnosis (< 30 x 10(9)/L vs > 30 x 10(9)/L) and disease status (CR1 vs > CR1). The median time to relapse for CR1 and for beyond CR1 patients was 12 months and 9 months, respectively. Our results indicate that FTBI/VP16 with or without cyclophosphamide confers long-term survival in Ph(+) ALL patients and that disease status at the time of HCT is an important predictor of outcome.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Benzamidas , Niño , Preescolar , Terapia Combinada , Fraccionamiento de la Dosis de Radiación , Etopósido/uso terapéutico , Femenino , Estudios de Seguimiento , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/mortalidad , Humanos , Mesilato de Imatinib , Masculino , Persona de Mediana Edad , Piperazinas/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Pirimidinas/uso terapéutico , Inducción de Remisión/métodos , Factores de Riesgo , Hermanos , Análisis de Supervivencia , Trasplante Homólogo , Resultado del Tratamiento , Irradiación Corporal Total
16.
Biol Blood Marrow Transplant ; 15(11): 1407-14, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19822300

RESUMEN

Acute lymphoblastic leukemia (ALL) with high-risk features has a poor prognosis in adults despite aggressive chemotherapy. Reduced-intensity conditioning (RIC) is a lower toxicity alternative for high-risk patients requiring hematopoietic cell transplantation (HCT); however, it has not been widely used for ALL. We conducted a retrospective study of 24 high-risk adult ALL patients who received an RIC regimen of fludarabine (Flu)/melphalan (Mel) prior to allogeneic peripheral blood stem cell transplantation (PBSCT) between 6/14/02 and 6/15/07 at the City of Hope. Indications for the RIC regimen were: (1) aged 50 years or older (42%), (2) compromised organ function (54%), or (3) recipient of a previous HCT (37.5%). Patients had a median age of 47.5 years and the median follow-up was 28.5 months for living patients. Both overall survival (OS) and disease-free survival (DFS) at 2 years was 61.5%. Relapse incidence was 21.1% and nonrelapse mortality (NRM) was 21.5% at 2 years. Chronic graft-versus-host (cGVHD) developed in 86% of evaluable patients. In this series, no significant correlations were made between outcomes and patient age, presence of Philadelphia chromosome, relatedness of donor source, or prior HCT. These high survival rates for high-risk ALL patients following RIC HCT may offer a promising option for patients not eligible for a standard myeloablative transplant.


Asunto(s)
Melfalán/administración & dosificación , Agonistas Mieloablativos/administración & dosificación , Trasplante de Células Madre de Sangre Periférica , Leucemia-Linfoma Linfoblástico de Células Precursoras/cirugía , Acondicionamiento Pretrasplante/métodos , Vidarabina/análogos & derivados , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Benzamidas , Terapia Combinada , Dasatinib , Supervivencia sin Enfermedad , Esquema de Medicación , Femenino , Estudios de Seguimiento , Supervivencia de Injerto , Enfermedad Injerto contra Huésped/epidemiología , Humanos , Mesilato de Imatinib , Masculino , Persona de Mediana Edad , Neoplasias Primarias Secundarias/tratamiento farmacológico , Neoplasias Primarias Secundarias/epidemiología , Neoplasias Primarias Secundarias/cirugía , Piperazinas/administración & dosificación , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/epidemiología , Pirimidinas/administración & dosificación , Reoperación , Estudios Retrospectivos , Riesgo , Tiazoles/administración & dosificación , Trasplante Homólogo , Vidarabina/administración & dosificación , Adulto Joven
17.
Clin Cancer Res ; 14(10): 2927-35, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18483359

RESUMEN

PURPOSE: Gene copy number alteration (CNA) is common in malignant melanoma and is associated with tumor development and progression. The concordance between molecular cytogenetic techniques used to determine CNA has not been evaluated on a large set of loci in malignant melanoma. EXPERIMENTAL DESIGN: A panel of 16 locus-specific fluorescence in situ hybridization (FISH) probes located on eight chromosomes was used to identify CNA in touch preparations of frozen tissue samples from 19 patients with metastatic melanoma (SWOG-9431). A subset (n = 11) was analyzed using bacterial artificial chromosome (BAC) array comparative genomic hybridization (aCGH) of DNA isolated directly from touch-preparation slides. RESULTS: By FISH, most samples showed loss near or at WISP3/6p21, CCND3/6q22, and CDKN2A/9p21 (>75% of samples tested). More than one third of CDKN2A/9p21 losses were biallelic. Gains of NEDD9/6p24, MET/7q31, and MYC/8q24 were common (57%, 47%, and 41%, respectively) and CNA events involving 9p21/7p12.3 and MET were frequently coincident, suggesting gain of the whole chromosome 7. Changes were confirmed by aCGH, which also uncovered many discreet regions of change, larger than a single BAC. Overlapping segments observed in >45% of samples included many of the loci analyzed in the FISH study, in addition to other WNT pathway members, and genes associated with TP53 pathways and DNA damage response, repair, and stability. CONCLUSIONS: This study outlines a set of CNAs at the gene and regional level, using FISH and aCGH, which may provide a benchmark for future studies and may be important in selection of individual therapy for patients with metastatic malignant melanoma.


Asunto(s)
Análisis Citogenético , Dosificación de Gen , Perfilación de la Expresión Génica , Melanoma/genética , Perfilación de la Expresión Génica/métodos , Humanos , Hibridación Fluorescente in Situ , Análisis de Secuencia por Matrices de Oligonucleótidos , Reproducibilidad de los Resultados
18.
Leuk Res ; 32(9): 1329-32, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18423850

RESUMEN

The inaugural meeting of the International Working Group on MDS cytogenetics convened 22-23 October 2007 in Chicago, IL. Under the sponsorship of the Myelodysplastic Syndromes Foundation, the group was organized to address the substantial need for worldwide standardized cytogenetic testing for MDS in clinical practice and research. Eighteen cytogeneticists from 10 countries attended the first working group meeting. Representatives from France and Austria were unable to attend the Chicago meeting. Marilyn L. Slovak, PhD (City of Hope, USA) served as Working Group Chair and Gordon Dewald, PhD (Mayo Clinic, USA), served as Working Group Advisor and Co-Chair. Other members in attendance included: Mette Andersen, Rigshospitalet, Denmark; Lynda Campbell, St. Vincent's Hospital Melbourne, Australia; Athena Cherry, Stanford University, USA; Kathy Chun, North York General Hospital, Canada; Mike Griffiths, West Midlands Regional Genetics Lab, UK; Detlef Haase, Georg-August-Universität, Germany; Claudia Haferlach, MLL Münchner Leukämielabor GmbH, Germany; Anne Hagemeijer, University of Leuven, Belgium; Barbara Hildebrandt, Institut für Humangenetik & Anthropologie Dupsilonsseldorf, Germany; Douglas Horsman, BC Cancer Agency, Canada; M. Anwar Iqbal, University of Rochester, USA; Suresh Jhanwar, Memorial Sloan-Kettering Cancer Center, USA; Bertil Johansson, University Hospital, Sweden; Michelle LeBeau, University of Chicago, USA; Kazuma Ohyashiki, Tokyo Medical University, Japan; Francesc Solé, Hospital del Mar, Spain. The focus of the working group was to establish the natural history and clinical significance of cytogenetic anomalies associated with the myelodysplastic syndromes (MDS), and to incorporate cytogenetic testing into the development of new treatments to cure MDS. Three specific goals were discussed in an effort to rapidly improve the care of patients with MDS. The first goal was how to educate physicians on the appropriate use of cost effective cytogenetic testing for their patients with MDS. The second goal discussed was how best this working group could assist pharmaceutical companies with the use of appropriate cytogenetic testing in their evaluation of new drugs. The final goal discussed was how to advance cytogenetic research into the origin, progression and clinical significance of genetic anomalies associated with MDS.


Asunto(s)
Análisis Citogenético/métodos , Cooperación Internacional , Síndromes Mielodisplásicos/diagnóstico , Ensayos Clínicos como Asunto , Humanos , Síndromes Mielodisplásicos/terapia
19.
Leuk Res ; 32(11): 1770-5, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18417213

RESUMEN

Acute megakaryocytic leukemia is a rare form of acute myelogenous leukemia and may occur either de novo or by transformation of a preexisting myelodysplastic or myeloproliferative process including blast crisis of chronic myeloid leukemia (CML). Megakaryocytic blast crisis as the presenting manifestation of CML is extremely rare. We describe such a patient with no prior hematologic disease who presented with acute megakaryoblastic leukemia and extramedullary involvement, in whom the leukemic cells carried the BCR-ABL1 translocation as part of a complex karyotype. Using targeted sequential fluorescence in situ hybridization (T-FISH) technique, we detected two copies of BCR-ABL1 fusion gene in the leukemic blasts while the neutrophils carried a single copy of BCR-ABL1 fusion gene, thereby proving the origin of the megakaryoblastic leukemia from a previously undiagnosed CML clone. Blast crisis as a presenting manifestation of CML is rare and detecting clonal evolution of acute leukemia by specialized cytogenetic techniques may have important diagnostic and therapeutic implications.


Asunto(s)
Crisis Blástica/diagnóstico , Proteínas de Fusión bcr-abl/genética , Leucemia Megacarioblástica Aguda/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/diagnóstico , Megacariocitos/patología , Crisis Blástica/genética , Crisis Blástica/terapia , Humanos , Hibridación Fluorescente in Situ , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Masculino , Persona de Mediana Edad , Translocación Genética
20.
Cancer Genet ; 228-229: 218-235, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30344013

RESUMEN

Structural genomic abnormalities, including balanced chromosomal rearrangements, copy number gains and losses and copy-neutral loss-of-heterozygosity (CN-LOH) represent an important category of diagnostic, prognostic and therapeutic markers in acute myeloid leukemia (AML). Genome-wide evaluation for copy number abnormalities (CNAs) is at present performed by karyotype analysis which has low resolution and is unobtainable in a subset of cases. Furthermore, examination for possible CN-LOH in leukemia cells is at present not routinely performed in the clinical setting. Chromosomal microarray (CMA) analysis is a widely available assay for CNAs and CN-LOH in diagnostic laboratories, but there are currently no guidelines how to best incorporate this technology into clinical testing algorithms for neoplastic diseases including AML. The Cancer Genomics Consortium Working Group for Myeloid Neoplasms performed an extensive review of peer-reviewed publications focused on CMA analysis in AML. Here we summarize evidence regarding clinical utility of CMA analysis in AML extracted from published data, and provide recommendations for optimal utilization of CMA testing in the diagnostic workup. In addition, we provide a list of CNAs and CN-LOH regions which have documented clinical significance in diagnosis, prognosis and treatment decisions in AML.


Asunto(s)
Variaciones en el Número de Copia de ADN , Medicina Basada en la Evidencia , Leucemia Mieloide Aguda/genética , Pérdida de Heterocigocidad , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA