Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
ESMO Open ; 7(2): 100403, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35272130

RESUMEN

BACKGROUND: The COVID-19 pandemic has created enormous challenges for the clinical management of patients with hematological malignancies (HMs), raising questions about the optimal care of this patient group. METHODS: This consensus manuscript aims at discussing clinical evidence and providing expert advice on statements related to the management of HMs in the COVID-19 pandemic. For this purpose, an international consortium was established including a steering committee, which prepared six working packages addressing significant clinical questions from the COVID-19 diagnosis, treatment, and mitigation strategies to specific HMs management in the pandemic. During a virtual consensus meeting, including global experts and lead by the European Society for Medical Oncology and the European Hematology Association, statements were discussed and voted upon. When a consensus could not be reached, the panel revised statements to develop consensual clinical guidance. RESULTS AND CONCLUSION: The expert panel agreed on 33 statements, reflecting a consensus, which will guide clinical decision making for patients with hematological neoplasms during the COVID-19 pandemic.


Asunto(s)
COVID-19 , Neoplasias Hematológicas , Humanos , Consenso , Prueba de COVID-19 , Neoplasias Hematológicas/epidemiología , Neoplasias Hematológicas/terapia , Pandemias
3.
Blood ; 133(10): 1020-1030, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30404811

RESUMEN

The heterogeneity of myelodysplastic syndromes (MDSs) has made evaluating patient response to treatment challenging. In 2006, the International Working Group (IWG) proposed a revision to previously published standardized response criteria (IWG 2000) for uniformly evaluating clinical responses in MDSs. These IWG 2006 criteria have been used prospectively in many clinical trials in MDSs, but proved challenging in several of them, especially for the evaluation of erythroid response. In this report, we provide rationale for modifications (IWG 2018) of these recommendations, mainly for "hematological improvement" criteria used for lower-risk MDSs, based on recent practical and reported experience in clinical trials. Most suggestions relate to erythroid response assessment, which are refined in an overall more stringent manner. Two major proposed changes are the differentiation between "procedures" and "criteria" for hematologic improvement-erythroid assessment and a new categorization of transfusion-burden subgroups.


Asunto(s)
Ensayos Clínicos como Asunto/normas , Hematología/métodos , Hematología/normas , Síndromes Mielodisplásicos/terapia , Transfusión Sanguínea , Linaje de la Célula , Progresión de la Enfermedad , Transfusión de Eritrocitos , Eritrocitos/citología , Humanos , Cooperación Internacional , Recuento de Leucocitos , Neutrófilos , Recuento de Plaquetas , Guías de Práctica Clínica como Asunto , Calidad de Vida , Recurrencia , Conducta de Reducción del Riesgo , Sociedades Médicas , Resultado del Tratamiento
6.
Leukemia ; 32(2): 413-418, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28607470

RESUMEN

Chronic myelomonocytic leukemia (CMML) is a complex clonal hematological disorder classified among myelodysplastic (MDS)/myeloproliferative neoplasms. Prognosis is poor and there is a lack of effective treatments. The hypomethylating agent decitabine has shown activity against MDS and elderly acute myeloid leukemia, but there is little data focusing specifically on its efficacy in CMML. In this prospective, phase 2 Italian study, CMML patients received intravenous decitabine 20 mg/m2 per day on Days 1-5 of a 28-day treatment cycle. Response was evaluated after four and six cycles; patients responding at the end of six cycles could continue treatment with decitabine. Forty-three patients were enrolled; >50% were high-risk according to four CMML-specific scoring systems. In the intent-to-treat population (n=42), the overall response rate after six cycles was 47.6%, with seven complete responses (16.6%), eight marrow responses (19%), one partial response (2.4%) and four hematological improvements (9.5%). After a median follow-up of 51.5 months (range: 44.4-57.2), median overall survival was 17 months, with responders having a significantly longer survival than non-responders (P=0.02). Grade 3/4 anemia, neutropenia and thrombocytopenia occurred in 28.6%, 50% and 38% of patients, respectively. Decitabine appears to be an effective and well-tolerated treatment for patients with high-risk CMML.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Decitabina/administración & dosificación , Leucemia Mielomonocítica Crónica/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Italia , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Leucemia Mielomonocítica Crónica/patología , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Resultado del Tratamiento
7.
J Thromb Haemost ; 15(12): 2388-2392, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28976612

RESUMEN

Essentials Thrombocytopenia 2 (THC2) is an inherited thrombocytopenia (IT) with dysmegakaryopoiesis. Physicians often do not suspect the genetic origin of thrombocytopenia in patients with THC2. We report two THC2 patients misdiagnosed with myelodysplasia and treated with chemotherapy. IT should be always considered in patients with isolated thrombocytopenia and dysmegakaryopoiesis. SUMMARY: Thrombocytopenia 2 (THC2) is an autosomal-dominant disorder caused by point substitutions in the 5'UTR of the ANKRD26 gene. Patients have congenital thrombocytopenia, normal platelet morphology and function, and dysmegakaryopoiesis. Thrombocytopenia is frequently discovered only in adulthood and physicians often do not suspect its genetic origin. We describe two unrelated patients referred to two different institutions for investigation of thrombocytopenia. Based on the finding of dysmegakaryopoiesis at bone marrow examination, patients were diagnosed with myelodysplastic syndrome (MDS) (refractory thrombocytopenia) and treated with several courses of 5-azacytidine. Subsequently, demonstration of thrombocytopenia in their relatives eventually led to molecular diagnosis of THC2 in both families. These cases highlight that patients with THC2 are at risk of being misdiagnosed with MDS and receiving undue myelosuppressive treatments. Because dysmegakaryopoiesis is a feature also of other forms of inherited thrombocytopenia, a genetic disorder must always be considered when a patient presents with isolated thrombocytopenia and dysmegakaryopoiesis.


Asunto(s)
Trastornos de los Cromosomas/diagnóstico , Trastornos de los Cromosomas/genética , Mutación , Síndromes Mielodisplásicos/diagnóstico , Proteínas Nucleares/genética , Trombocitopenia/congénito , Anciano , Médula Ósea/patología , Rotura Cromosómica , Trastornos de los Cromosomas/patología , Análisis Mutacional de ADN , Errores Diagnósticos , Predisposición Genética a la Enfermedad , Humanos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Persona de Mediana Edad , Trombocitopenia/diagnóstico , Trombocitopenia/genética , Trombocitopenia/patología , Trombopoyesis/genética
8.
Leukemia ; 31(11): 2449-2457, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28321120

RESUMEN

Allogeneic hematopoietic stem cell transplantation (allo-SCT) represents the only curative treatment for patients with myelodysplastic syndrome (MDS), but involves non-negligible morbidity and mortality. Crucial questions in clinical decision-making include the definition of optimal timing of the procedure and the benefit of cytoreduction before transplant in high-risk patients. We carried out a decision analysis on 1728 MDS who received supportive care, transplantation or hypomethylating agents (HMAs). Risk assessment was based on the revised International Prognostic Scoring System (IPSS-R). We used a continuous-time multistate Markov model to describe the natural history of disease and evaluate the effect of different treatment policies on survival. Life expectancy increased when transplantation was delayed from the initial stages to intermediate IPSS-R risk (gain-of-life expectancy 5.3, 4.7 and 2.8 years for patients aged ⩽55, 60 and 65 years, respectively), and then decreased for higher risks. Modeling decision analysis on IPSS-R versus original IPSS changed transplantation policy in 29% of patients, resulting in a 2-year gain in life expectancy. In advanced stages, HMAs given before transplant is associated with a 2-year gain-of-life expectancy, especially in older patients. These results provide a preliminary evidence to maximize the effectiveness of allo-SCT in MDS.


Asunto(s)
Técnicas de Apoyo para la Decisión , Trasplante de Células Madre Hematopoyéticas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Años de Vida Ajustados por Calidad de Vida
10.
Leukemia ; 30(5): 1133-42, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26710888

RESUMEN

DNA methyltransferase 3A (DNMT3A) mutations are observed in myeloid malignancies, including myeloproliferative neoplasms (MPN), myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Transplantation studies have elucidated an important role for Dnmt3a in stem cell self-renewal and in myeloid differentiation. Here, we investigated the impact of conditional hematopoietic Dnmt3a loss on disease phenotype in primary mice. Mx1-Cre-mediated Dnmt3a ablation led to the development of a lethal, fully penetrant MPN with myelodysplasia (MDS/MPN) characterized by peripheral cytopenias and by marked, progressive hepatomegaly. We detected expanded stem/progenitor populations in the liver of Dnmt3a-ablated mice. The MDS/MPN induced by Dnmt3a ablation was transplantable, including the marked hepatomegaly. Homing studies showed that Dnmt3a-deleted bone marrow cells preferentially migrated to the liver. Gene expression and DNA methylation analyses of progenitor cell populations identified differential regulation of hematopoietic regulatory pathways, including fetal liver hematopoiesis transcriptional programs. These data demonstrate that Dnmt3a ablation in the hematopoietic system leads to myeloid transformation in vivo, with cell-autonomous aberrant tissue tropism and marked extramedullary hematopoiesis (EMH) with liver involvement. Hence, in addition to the established role of Dnmt3a in regulating self-renewal, Dnmt3a regulates tissue tropism and limits myeloid progenitor expansion in vivo.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/fisiología , Células Madre Hematopoyéticas/citología , Células Mieloides/citología , Animales , Células de la Médula Ósea , Movimiento Celular , Proliferación Celular , Autorrenovación de las Células , ADN Metiltransferasa 3A , Hematopoyesis , Hígado/patología , Ratones
11.
Blood Cancer J ; 5: e347, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26383820

RESUMEN

The aim of this study was to investigate the effects of a non-standard, intermittent imatinib treatment in elderly patients with Philadelphia-positive chronic myeloid leukaemia and to answer the question on which dose should be used once a stable optimal response has been achieved. Seventy-six patients aged ⩾65 years in optimal and stable response with ⩾2 years of standard imatinib treatment were enrolled in a study testing a regimen of intermittent imatinib (INTERIM; 1-month on and 1-month off). With a minimum follow-up of 6 years, 16/76 patients (21%) have lost complete cytogenetic response (CCyR) and major molecular response (MMR), and 16 patients (21%) have lost MMR only. All these patients were given imatinib again, the same dose, on the standard schedule and achieved again CCyR and MMR or an even deeper molecular response. The probability of remaining on INTERIM at 6 years was 48% (95% confidence interval 35-59%). Nine patients died in remission. No progressions were recorded. Side effects of continuous treatment were reduced by 50%. In optimal and stable responders, a policy of intermittent imatinib treatment is feasible, is successful in about 50% of patients and is safe, as all the patients who relapsed could be brought back to optimal response.


Asunto(s)
Antineoplásicos/administración & dosificación , Mesilato de Imatinib/administración & dosificación , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Femenino , Humanos , Mesilato de Imatinib/efectos adversos , Hibridación Fluorescente in Situ , Estimación de Kaplan-Meier , Leucemia Mielógena Crónica BCR-ABL Positiva/mortalidad , Masculino , Proyectos Piloto , Inducción de Remisión/métodos
13.
Leukemia ; 28(3): 621-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24192812

RESUMEN

The nucleoside analog azacitidine (AZA) is used in the treatment of myelodysplastic syndromes (MDS), but 30-40% of patients fail to respond or relapse after treatment. Hence, to identify new molecular alterations that allow for identification of patients unlikely to respond to AZA could impact the utility of this therapy. We determined the expression levels of genes involved in AZA metabolism: UCK1, UCK2, DCK, hENT1, RRM1 and RRM2 using quantitative PCR in samples from 57 patients with MDS who received AZA. Lower expression of UCK1 was seen in patients without a response to AZA (median 0.2 vs 0.49 for patients with response to AZA, P=0.07). This difference in UCK1 expression was not influenced by aberrant methylation of the UCK1 promoter. In addition, the seven polymorphic loci found in the coding sequence were not associated with UCK1 gene expression nor AZA response. Silencing of UCK1 by siRNA leads to blunted response to AZA in vitro. The univariate analysis revealed that patients expressing lower than median levels of UCK1 had a shorter overall survival (P=0.049). Our results suggest that expression level of UCK1 is correlated with clinical outcome and may influence the clinical response to AZA treatment in patients with MDS.


Asunto(s)
Antineoplásicos/uso terapéutico , Azacitidina/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Enzimas/metabolismo , Síndromes Mielodisplásicos/enzimología , Nucleósidos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Enzimas/genética , Femenino , Silenciador del Gen , Humanos , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/tratamiento farmacológico , Reacción en Cadena de la Polimerasa , Pronóstico
16.
Leuk Res ; 34(12): 1576-88, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20149927

RESUMEN

Since 2002, date of publication of the previous Italian Society of Haematology (SIE) practice guidelines for management of myelodysplastic syndromes (MDS), novel disease-modifying treatments have been introduced and the SIE commissioned an update. After a comprehensive review of the medical literature published since January 2001, the Expert Panel formulated recommendations for the management of adult and paediatric MDS, graded according to the available evidence. The major updates are: first-line hypomethylating agents in patients with INT2-high-risk disease; controlled use of first-line lenalidomide in low-INT1 risk transfusion-dependent patients with 5q deletion; deferasirox in low-INT1 patients with a relevant transfusional load; first-line high-dose ESA in low-INT1 patients with Hb <10 g/dl and endogenous EPO <500 U/l; allogeneic HSCT first-line therapy for INT2- and high-risk patients <65 years without severe co morbidities.


Asunto(s)
Síndromes Mielodisplásicos/terapia , Adolescente , Adulto , Antineoplásicos/uso terapéutico , Niño , Preescolar , Humanos , Lenalidomida , Persona de Mediana Edad , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/mortalidad , Talidomida/análogos & derivados , Talidomida/uso terapéutico
17.
J Chemother ; 20(3): 291-6, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18606581

RESUMEN

Myelodysplastic syndromes (MDS) are very heterogeneous diseases in terms of clinical presentation and prognosis. Patients with pure red cell dysplasias have a life expectancy of more than 10 years, whereas those with refractory anemias with excess blasts have survival shorter than 6 months. Until a few years ago, therapeutic options were palliative and supportive care only. Quite recently, the treatment panorama for MDS has radically changed and the different biological behavior of MDS requires a precise choice among completely different therapies: immunosuppressive agents, anti-apoptotic agents and growth factors are effective in low risk MDS, whereas epigenetic drugs, tyrosine kinase inhibitors, and possibly high dose chemotherapy and bone marrow transplantation are valuable in high risk MDS. We review the results of such therapies and the selection criteria for MDS patients.


Asunto(s)
Síndromes Mielodisplásicos/tratamiento farmacológico , Selección de Paciente , Inhibidores Enzimáticos/uso terapéutico , Inhibidores de Histona Desacetilasas , Humanos , Inmunosupresores/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Síndromes Mielodisplásicos/terapia , Trasplante de Células Madre
18.
Ecancermedicalscience ; 2: 121, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-22275991

RESUMEN

Azacitidine (AZA), as demonstrated in the phase III trial (AZA-001), is the first MDS treatment to significantly prolong overall survival (OS) in higher risk MDS pts ((2007) Blood 110 817). Approximately, one-third of the patients (pts) enrolled in AZA-001 were FAB RAEB-T (≥20-30% blasts) and now meet the WHO criteria for acute myeloid leukaemia (AML) ((1999) Blood 17 3835). Considering the poor prognosis (median survival <1 year) and the poor response to chemotherapy in these pts, this sub-group analysis evaluated the effects of AZA versus conventional care regimens (CCR) on OS and on response rates in pts with WHO AML.

19.
Ecancermedicalscience ; 2: 118, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-22275990

RESUMEN

The international, phase III, multi-centre AZA-001 trial demonstrated azacitidine (AZA) is the first treatment to significantly extend overall survival (OS) in higher risk myelodysplastic syndromes (MDS) patients (Fenaux (2007) Blood110 817). The current treatment paradigm, which is based on a relationship between complete remission (CR) and survival, is increasingly being questioned (Cheson (2006) Blood108 419). Results of AZA-001 show CR is sufficient but not necessary to prolong OS (List (2008) Clin Oncol26 7006). Indeed, the AZA CR rate in AZA-001 was modest (17%), while partial remission (PR, 12%) and haematological improvement (HI, 49%) were also predictive of prolonged survival. This analysis was conducted to assess the median number of AZA treatment cycles associated with achievement of first response, as measured by IWG 2000-defined CR, PR or HI (major + minor). The number of treatment cycles from first response to best response was also measured.

20.
Oncogene ; 27(12): 1767-78, 2008 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-17891169

RESUMEN

We analysed the in vitro effects of a new hydroxamate derivative, ITF2357, on AML cells. ITF2357 potently induced histone acetylation. ITF2357 0.1 microM blocked proliferation and induced apoptosis in AML1/ETO-positive Kasumi-1 cells, while AML1/ETO-negative HL60, THP1 and NB4 cell lines were sensitive only to 1 microM ITF2357. Apoptosis was induced by 0.1 microM ITF2357 in AML1/ETO-positive primary blasts and U937-A/E cells induced to express AML1/ETO, but not in U937-A/E cells non-expressing AML1/ETO. In Kasumi-1 cells 0.1 microM ITF2357 induced AML1/ETO degradation through a caspase-dependent mechanism. ITF2357 0.1 microM also determined DNMT1 efflux from, and p300 influx to, the nucleus. Moreover, 0.1 microM ITF2357 determined local H4 acetylation and release of DNMT1, HDAC1 and AML1/ETO, paralleled by recruitment of p300 to the IL-3 gene promoter. ITF2357 treatment, however, did not induce re-expression of IL-3 gene. Accordingly, the methylation level of IL-3 promoter, as well as of several other genes, was unmodified. In conclusion, ITF2357 emerged as an anti-leukaemic agent very potent on AML cells, and on AML1/ETO-positive cells in particular. More relevantly, clearly emerged from our results that ITF2357 could be an ideal agent to treat AML subtypes presenting AML1/ETO fusion protein which determine HDAC involvement in leukaemogenesis.


Asunto(s)
Antineoplásicos/farmacología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/farmacología , Leucemia/tratamiento farmacológico , Proteínas Proto-Oncogénicas/biosíntesis , Factores de Transcripción/biosíntesis , Acetilación , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Células HL-60 , Humanos , Leucemia/enzimología , Leucemia/patología , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/enzimología , Proteína 1 Compañera de Translocación de RUNX1 , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...