Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Br J Haematol ; 192(5): 879-891, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33486765

RESUMEN

Ineffective erythropoiesis and iron overload are common in myelodysplastic syndromes (MDS). Erythroferrone (ERFE) and growth/differentiation factor 15 (GDF15) are two regulators of iron homeostasis produced by erythroid progenitors. Elevated systemic levels of ERFE and GDF15 in MDS are associated with dysregulated iron metabolism and iron overload, which is especially pronounced in MDS with SF3B1 gene mutations. However, the role of ERFE and GDF15 in MDS pathogenesis and their influence on disease progression are largely unknown. Here, we analyzed the expression of ERFE and GDF15 in CD71+ erythroid progenitors of n = 111 MDS patients and assessed their effects on patient survival. The expression of ERFE and GDF15 in MDS was highly aberrant. Unexpectedly, ERFE expression in erythroprogenitors was highly relevant for MDS prognosis and independent of International Prognostic Scoring System (IPSS) stratification. Although ERFE expression was increased in patients with SF3B1 mutations, it predicted overall survival (OS) in both the SF3B1wt and SF3B1mut subgroups. Of note, ERFE overexpression predicted superior OS in the IPSS low/Int-1 subgroup and in patients with normal karyotype. Similar observations were made for GDF15, albeit not reaching statistical significance. In summary, our results revealed a strong association between ERFE expression and MDS outcome, suggesting a possible involvement of ERFE in molecular MDS pathogenesis.


Asunto(s)
Antígenos CD/análisis , Células Precursoras Eritroides/metabolismo , Síndromes Mielodisplásicos/metabolismo , Hormonas Peptídicas/biosíntesis , Receptores de Transferrina/análisis , Adulto , Anciano , Anciano de 80 o más Años , Células Precursoras Eritroides/química , Femenino , Factor 15 de Diferenciación de Crecimiento/biosíntesis , Factor 15 de Diferenciación de Crecimiento/genética , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/mortalidad , Síndromes Mielodisplásicos/terapia , Hormonas Peptídicas/genética , Fosfoproteínas/genética , Modelos de Riesgos Proporcionales , Factores de Empalme de ARN/genética , Resultado del Tratamiento , Adulto Joven
2.
J Cancer Res Clin Oncol ; 144(8): 1531-1538, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29761371

RESUMEN

BACKGROUND: Iron overload (IOL) due to repetitive transfusions of packed red blood cells (pRBC) has a major impact on morbidity and mortality in patients with inherited bone marrow failure syndromes and hemoglobinopathies such as thalassemia and sickle cell disease. However, whether IOL influences the outcome of elderly patients with myeloid malignancies is not yet clear. Moreover, clinical trials have reported high drop-out rates during treatment with the oral iron chelator deferasirox (DFX). AIM: Here we report the results of a 2-year prospective observational study that aimed at describing the routine use of DFX in patients with hematological malignancies with regard to safety, efficacy and handling of the drug in a routine setting. RESULTS: A total of 406 patients were included. 58% of the patients were male. Most of the patients had myelodysplastic syndromes (MDS) (68%) and myeloproliferative neoplasms (MPN) (14%). Median time from first transfusion to study enrollment was 1.1 years (0-25.5 years) and most patients were chelation naive (91%) at enrollment. With regard to transfusion burden, most of the patients were moderately or mildly transfusion-dependent with 53% receiving 2-4 and 27% receiving less than 2 units of pRBC per month. Serum ferritin decreased from a mean of 2305 µg/l (± 1449 µg/l) to a mean of 1910 µg/l (± 1529 µg/l) at 24 months. There was no substantial change in transfusion-dependence during the observation period. Dose adjustments were reported in 48% of the patients with dose-escalation strategies being the most frequent reason for dosage increases (49%). The median observation time was 355 days (5-1080 days). Median duration of exposure to DFX was 322 days (2-1078 days). Two-hundred and ninety (72%) patients discontinued the trial prematurely after a median time of 235 days (1-808 days). Death (29%) and adverse events (23%) were the main reasons for discontinuation. Eleven percent of the patients discontinued treatment due to sufficient decrease in serum ferritin. Most frequent adverse events were decrease in creatinine clearance (22%), increase in serum creatinine (18%) and diarrhea (16%). CONCLUSION: This descriptive trial confirms the efficacy of DFX in decreasing the serum ferritin. Moreover, the high drop-out rates seen in prospective trials are recapitulated in this study, which can be attributed to adverse events in a substantial proportion of patients.


Asunto(s)
Deferasirox/uso terapéutico , Transfusión de Eritrocitos/efectos adversos , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Femenino , Alemania , Enfermedades Hematológicas/terapia , Humanos , Sobrecarga de Hierro/etiología , Masculino , Persona de Mediana Edad , Adulto Joven
3.
Leuk Res ; 68: 62-69, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29549809

RESUMEN

In low risk MDS, increased apoptosis of erythroid progenitors mediated via CD95 (Fas) activation has been described to result in peripheral cytopenia. Blockade of the CD95 system can improve erythropoiesis in MDS. Asunercept (APG101) is a fusion protein consisting of the extracellular domain of human CD95 and the Fc domain of human IgG1 blocking the interaction between CD95 and its ligand. Here we report on results from a phase I study in 20 transfusion-dependent low and intermediate risk MDS patients treated with intravenous asunercept (EudraCT 2012-003027-37). Primary objectives were safety and tolerability as well as pharmacodynamic effects. Secondary objectives were hematologic improvement, incidence and time to leukemic progression as well as overall survival. Frequency and severity of adverse events were in range of what could be expected in a patient cohort comprising of elderly MDS patients. Two patients experienced a serious adverse event with a suspected relationship to asunercept. The incidence of disease progression was low. In the 20 patients a decrease of the transfusion need from a mean of 10,8 (±5,1) pRBCs during the 12 weeks treatment phase to a mean of 10,0 (±4,2) pRBCs at the end of the study was observed. In conclusion, asunercept was well tolerated and showed efficacy in transfusion-dependent low and intermediate risk MDS patients. Further clinical investigation is warranted, particularly in combination with erythropoiesis stimulating agents (ESAs).


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Transfusión Sanguínea , Inmunoglobulina G/efectos adversos , Inmunoglobulina G/uso terapéutico , Síndromes Mielodisplásicos/tratamiento farmacológico , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/uso terapéutico , Receptor fas/uso terapéutico , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Proteína Ligando Fas/antagonistas & inhibidores , Femenino , Humanos , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/farmacología , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/patología , Estudios Prospectivos , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/farmacología , Riesgo , Receptor fas/administración & dosificación
4.
Ann Hematol ; 92(10): 1325-33, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23645217

RESUMEN

Centrosomes play important roles in the maintenance of genetic stability and centrosomal aberrations are a hallmark of cancer. Deregulation of centriole duplication leads to supernumerary centrosomes, sister chromatid missegregation and could result in chromosomal instability (CIN) and aneuploidy. CIN is a common feature in at least 45% of patients with myelodysplastic syndromes (MDS). Therefore, we sought to investigate the centrosomal status and its role for development of CIN in bone marrow (BM) cells of MDS patients. BM cells of 34 MDS patients were examined cytogenetically. Furthermore, cells were immunostained with a centrosome-specific antibody to pericentrin to analyze the centrosomal status. Umbilical cord blood specimens and BM cells of healthy persons (n = 11 and n = 4) served as controls. In addition, the protein expression of the protease separase responsible for genetic stability was examined by western blot analysis. Centrosome abnormalities were detected in 10% (range, 4-17%) of cells of MDS samples, but in only 2% (range, 0-4%) of cells of healthy controls. Normal karyotypes were found in control cells and in BM cells of 16/34 MDS patients. The incidence of centrosomal alterations was higher in BM cells of patients with cytogenetic alterations (mean, 12%) compared to BM cells of patients without cytogenetic changes (mean, 7%). Our results indicate that centrosome alterations are a common and early detectable feature in MDS patients and may contribute to the acquisition of chromosomal aberrations. We assume that centrosome defects could be involved in disease progression and may serve as a future prognostic marker.


Asunto(s)
Células de la Médula Ósea/metabolismo , Centrosoma/metabolismo , Inestabilidad Cromosómica/genética , Síndromes Mielodisplásicos/genética , Anciano , Femenino , Humanos , Cariotipo , Masculino , Persona de Mediana Edad
5.
J Cancer Res Clin Oncol ; 138(6): 953-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22358300

RESUMEN

BACKGROUND: Myelodysplastic syndromes (MDS) are mainly a disease of the elderly. Commonly, MDS patients are treated in an outpatient setting making hematological/oncological private practices (PP) an important backbone in the management of MDS patients. METHODS: To gain more insights into the characteristics of patients with MDS treated in hematological/oncological PP and to evaluate the daily diagnostic routines and classification systems used, we performed questionnaire-based analyses. Moreover, to investigate whether characteristics of MDS in PP differ from patients treated in specialized MDS centers in university hospitals (UH), we compared both cohorts of MDS patients. RESULTS: In total, 197 patients in PP and 165 patients in UH were enrolled. Patients in UH were significantly younger as compared to PP. Furthermore, in UH, a greater proportion of patients with international prognostic scoring system (IPSS) higher risk were found, whereas patients with IPSS lower risk were more frequent in PP. In addition, patients in UH had significantly lower hemoglobin levels and platelet counts compared to PP. CONCLUSION: Our data show that PP and UH are approached by different MDS patient cohorts resulting in different diagnostic workups of MDS patients.


Asunto(s)
Síndromes Mielodisplásicos/clasificación , Síndromes Mielodisplásicos/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Femenino , Alemania , Hospitales Universitarios , Humanos , Masculino , Persona de Mediana Edad , Práctica Privada , Pronóstico , Factores de Riesgo , Encuestas y Cuestionarios , Adulto Joven
6.
Leuk Res ; 35(9): 1131-5, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21737138

RESUMEN

Myelodysplastic syndromes are characterized by ineffective hematopoiesis resulting in peripheral cytopenias. The majority of patients is dependent on regular transfusions of packed red blood cells leading to a secondary iron overload which might result in organ damage. Therefore, sufficient iron chelation therapy in selected patients is mandatory. Deferasirox (DFX) is an orally administered iron chelator which has been highly efficient in the treatment of secondary iron overload. Most frequent side effects of DFX are gastrointestinal disturbances, which leads in some patients to low adherence to the therapy. An expert panel met in Lisbon in July 2010 to develop recommendations on prevention and management of GI disturbances based on existing data and personal experiences.


Asunto(s)
Benzoatos/efectos adversos , Benzoatos/uso terapéutico , Enfermedades Gastrointestinales/inducido químicamente , Enfermedades Gastrointestinales/terapia , Síndromes Mielodisplásicos/tratamiento farmacológico , Triazoles/efectos adversos , Triazoles/uso terapéutico , Terapia por Quelación/efectos adversos , Terapia por Quelación/métodos , Deferasirox , Enfermedades Gastrointestinales/prevención & control , Humanos , Quelantes del Hierro/efectos adversos , Quelantes del Hierro/uso terapéutico , Síndromes Mielodisplásicos/complicaciones , Guías de Práctica Clínica como Asunto
7.
J Clin Oncol ; 29(18): 2499-506, 2011 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-21576631

RESUMEN

PURPOSE: To study the incidence and prognostic impact of mutations in Additional sex comb-like 1 (ASXL1) in a large cohort of patients with myelodysplastic syndrome (MDS). PATIENTS, MATERIALS, AND METHODS: Overall, 193 patients with MDS and 65 healthy volunteers were examined for ASXL1 mutations by direct sequencing and for expression levels of ASXL1. The prognostic impact of ASXL1 mutation and expression levels was evaluated in the context of other clinical and molecular prognostic markers. RESULTS: Mutations in ASXL1 occurred with a frequency of 20.7% in MDS (n = 40 of 193) with 70% (n = 28) of mutations being frameshift mutations and 30% (n = 12) being heterozygous point mutations leading to translational changes. ASXL1 mutations were correlated with an intermediate-risk karyotype (P = .002) but not with other clinical parameters. The presence of ASXL1 mutations was associated with a shorter overall survival for frameshift and point mutations combined (hazard ratio [HR], 1.744; 95% CI, 1.08 to 2.82; P = .024) and for frameshift mutations only (HR, 2.06; 95% CI, 1.21 to 3.50; P = .008). ASXL1 frameshift mutations were associated with a reduced time to progression of acute myeloid leukemia (AML; HR 2.35; 95% CI, 1.17 to 4.74; P = .017). In multivariate analysis, when considering karyotype, transfusion dependence, and IDH1 mutation status, ASXL1 frameshift mutations remained an independent prognostic marker in MDS (overall survival: HR, 1.85; 95% CI, 1.03 to 3.34; P = .040; time to AML progression: HR, 2.39; 95% CI, 1.12 to 5.09; P = .024). CONCLUSION: These results suggest that ASXL1 mutations are frequent molecular aberrations in MDS that predict an adverse prognostic outcome. Screening of patients for ASXL1 mutations might be useful for clinical risk stratification and treatment decisions in the future.


Asunto(s)
Mutación del Sistema de Lectura , Síndromes Mielodisplásicos/genética , Mutación Puntual , Proteínas Represoras/análisis , Adulto , Anciano , Anciano de 80 o más Años , Suero Antilinfocítico , Biomarcadores , Ensayos Clínicos como Asunto/estadística & datos numéricos , Metilación de ADN , Análisis Mutacional de ADN , Progresión de la Enfermedad , Exones/genética , Femenino , Estudios de Seguimiento , Perfilación de la Expresión Génica , Genes MHC Clase II , Humanos , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/epidemiología , Masculino , Persona de Mediana Edad , Estudios Multicéntricos como Asunto/estadística & datos numéricos , Síndromes Mielodisplásicos/mortalidad , Síndromes Mielodisplásicos/terapia , Pronóstico , Modelos de Riesgos Proporcionales , ARN Mensajero/biosíntesis , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Riesgo
8.
Leuk Res ; 34(12): 1610-6, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20471677

RESUMEN

Secreted frizzled related protein 1 (SFRP1) is an extracellular antagonist of the Wnt signalling pathway that plays an important role in the pathogenesis of solid tumours and haematopoietic malignancies. SFRP1 has been observed to be transcriptionally down-regulated due to hypermethylation in acute and chronic leukaemia, but so far not in myelodysplastic syndrome (MDS). Moreover, it has been shown that the epigenetic inactivation of SFRP1 correlates with an overexpression of the Wnt receptor Frizzled 3 (Fzd3) in acute leukaemia. Using real-time quantitative reverse transcription polymerase chain reaction (RT-PCR) we examined mRNA expression of SFRP1 and Fzd3 in bone marrow cells derived from 121 patients with different risk types of MDS, acute myeloid leukaemia (AML) and acute lymphoblastic leukaemia (ALL). We employed pyrosequencing to quantify promoter DNA methylation in MDS and acute leukaemia. We detected significant lower mRNA transcription of SFRP1 in MDS compared to healthy individuals. However, DNA sequence mutations or frequent elevated DNA methylation levels of the SFRP1 promoter could not be observed in MDS but in AML and ALL as previously reported. The expression levels of Fzd3 were up-regulated in both acute leukaemia and MDS. Our data show a significant transcriptional down-regulation of SFRP1 as a common event in AML, ALL and - as demonstrated for the first time - in MDS. An inactivation of SFRP1 and the transcriptional up-regulation of Fzd3 seem to be associated with an activation of the Wnt signalling pathway in these haematopoietic diseases.


Asunto(s)
Metilación de ADN , Regulación hacia Abajo , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Proteínas de la Membrana/biosíntesis , Síndromes Mielodisplásicos/metabolismo , Regiones Promotoras Genéticas , Transcripción Genética , Enfermedad Aguda , Enfermedad Crónica , Femenino , Receptores Frizzled/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Síndromes Mielodisplásicos/patología , Receptores Acoplados a Proteínas G/metabolismo , Factores de Riesgo , Proteínas Wnt/antagonistas & inhibidores
9.
Haematologica ; 95(10): 1668-74, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20494930

RESUMEN

BACKGROUND: Myelodysplastic syndromes are a heterogeneous group of hematopoietic stem cell disorders with a high propensity to transform into acute myeloid leukemia. Heterozygous missense mutations in IDH1 at position R132 and in IDH2 at positions R140 and R172 have recently been reported in acute myeloid leukemia. However, little is known about the incidence and prognostic impact of IDH1 and IDH2 mutations in myelodysplastic syndromes. DESIGN AND METHODS: We examined 193 patients with myelodysplastic syndromes and 53 patients with acute myeloid leukemia arising from myelodysplastic syndromes for mutations in IDH1 (R132), IDH2 (R172 and R140), and NPM1 by direct sequencing. RESULTS: We found that mutations in IDH1 occurred with a frequency of 3.6% in myelodysplastic syndromes (7 mutations in 193 patients) and 7.5% in acute myeloid leukemia following myelodysplastic syndromes (4 mutations in 53 patients). Three mutations in codon R140 of IDH2 and one mutation in codon R172 were found in patients with acute myeloid leukemia following myelodysplastic syndromes (7.5%). No IDH2 R140 or R172 mutations were identified in patients with myelodysplastic syndromes. The presence of IDH1 mutations was associated with a shorter overall survival (HR 3.20; 95% CI 1.47-6.99) and a higher rate of transformation into acute myeloid leukemia (67% versus 28%, P=0.04). In multivariate analysis when considering karyotype, transfusion dependence and International Prognostic Scoring System score, IDH1 mutations remained an independent prognostic marker in myelodysplastic syndromes (HR 3.57; 95% CI 1.59-8.02; P=0.002). CONCLUSIONS: These results suggest that IDH1 mutations are recurrent molecular aberrations in patients with myelodysplastic syndromes, and may become useful as a poor risk marker in these patients. These findings await validation in prospective trials.


Asunto(s)
Isocitrato Deshidrogenasa/genética , Mutación , Síndromes Mielodisplásicos/genética , Biomarcadores , Análisis Mutacional de ADN , Humanos , Síndromes Mielodisplásicos/diagnóstico , Nucleofosmina , Pronóstico
10.
Blood ; 101(7): 2529-33, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12517808

RESUMEN

Alterations in hemostasis leading to symptomatic thromboembolism have been observed in patients with acute lymphoblastic leukemia (ALL) receiving Escherichia coli asparaginase (CASP) combined with steroids. Moreover, hereditary prothrombotic risk factors are associated with an increased risk for venous thromboembolism in pediatric ALL patients treated according to the BFM 90/95 protocols (including CASP combined with prednisone during induction therapy). To assess whether the thromboembolic risk associated with established prothrombotic risk factors is modified by treatment modalities (prednisone or dexamethasone), the present analysis was performed. Three hundred thirty-six consecutively recruited leukemic children treated according to different BFM protocols (PRED group, n = 280, 60 mg/m(2) prednisone; DEXA group, n = 56, 10 mg/m(2) dexamethasone during induction therapy) were studied. Study end point was the onset of symptomatic vascular accidents during induction therapy. Cumulative thromboembolism-free survival was significantly reduced in children in the PRED group (thrombosis frequency, 10.4%) compared with children in the DEXA group (thrombosis frequency, 1.8%; P =.028). Although no significant difference was found in the overall prevalence of prothrombotic risk factors, 46.5% of patients in the PRED group who experienced thromboembolic events were carriers of a prothrombotic risk factor, whereas no carrier in the DEXA group had a thromboembolism. At the time of maximum CASP activity, fibrinogen and activities of antithrombin, plasminogen, and protein S were significantly reduced in the PRED group. No significant correlation could be found between CASP activity and levels of coagulation factors. In conclusion, the use of dexamethasone instead of prednisone, administered with CASP, significantly reduced the onset of venous thromboembolism.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Asparaginasa/uso terapéutico , Daunorrubicina/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/sangre , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Prednisona/uso terapéutico , Vincristina/uso terapéutico , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Asparaginasa/administración & dosificación , Asparaginasa/efectos adversos , Niño , Preescolar , Daunorrubicina/administración & dosificación , Daunorrubicina/efectos adversos , Dexametasona/administración & dosificación , Humanos , Lactante , Masculino , Prednisona/administración & dosificación , Prednisona/efectos adversos , Factores de Riesgo , Tromboembolia/inducido químicamente , Tromboembolia/epidemiología , Vincristina/administración & dosificación , Vincristina/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...