Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Acta Haematol ; 147(4): 427-434, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38104534

RESUMEN

BACKGROUND: Most patients with lower risk myelodysplastic neoplasms or syndromes (MDSs) become RBC transfusion-dependent, resulting in iron overload, which is associated with an increased oxidative stress state. Iron-chelation therapy is applied to attenuate the toxic effects of this state. Deferiprone (DFP) is an oral iron chelator, which is not commonly used in this patient population, due to safety concerns, mainly agranulocytosis. The purpose of this study was to assess the effect of DFP, on oxidative stress parameters in iron-overloaded RBC transfusion-dependent patients with lower risk MDSs. METHODS: Adult lower risk MDS patients with a cumulative transfusion burden of >20 red blood cell units and evidence of iron overload (serum ferritin >1,000 ng/mL) were included in this study. DFP was administered (100 mg/kg/day) for 4 months. Blood samples for oxidative stress parameters and iron overload parameters were done at baseline and monthly: reactive oxygen species (ROS), phosphatidylserine, reduced glutathione, membrane lipid peroxidation, serum ferritin, and cellular labile iron pool. The primary efficacy variable was ROS. Tolerability and side effects were recorded as well. A paired t test was applied for statistical analyses. RESULTS: Eighteen patients were treated with DFP. ROS significantly decreased in all cell lineages: median decrease of 58.6% in RBC, 33.3% in PMN, and 39.8% in platelets (p < 0.01 for all). Other oxidative stress markers improved: phosphatidylserine decreased by 57.95%, lipid peroxidase decreased by 141.3%, and reduced gluthathione increased by 72.8% (p < 0.01 for all). The iron-overload marker and cellular labile iron pool decreased by 35% in RBCs, 44.3% in PMN, and 46.3% in platelets (p < 0.01 for all). No significant changes were observed in SF levels. There were no events of agranulocytosis. All AEs were grades 1-2. CONCLUSIONS: Herein, we showed preliminary evidence that DFP decreases iron-induced oxidative stress in MDS patients with a good tolerability profile (albeit a short follow-up period). No cases of severe neutropenia or agranulocytosis were reported. The future challenge is to prove that reduction in iron toxicity will eventually be translated into a clinically meaningful improvement.


Asunto(s)
Deferiprona , Quelantes del Hierro , Sobrecarga de Hierro , Síndromes Mielodisplásicos , Estrés Oxidativo , Humanos , Deferiprona/uso terapéutico , Deferiprona/farmacología , Estrés Oxidativo/efectos de los fármacos , Quelantes del Hierro/uso terapéutico , Quelantes del Hierro/farmacología , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/metabolismo , Masculino , Femenino , Anciano , Persona de Mediana Edad , Piridonas/uso terapéutico , Piridonas/efectos adversos , Piridonas/administración & dosificación , Anciano de 80 o más Años , Adulto , Israel , Administración Oral , Especies Reactivas de Oxígeno/metabolismo , Transfusión de Eritrocitos , Ferritinas/sangre
2.
Hematology ; 26(1): 88-95, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34000978

RESUMEN

Objectives: Reactive oxygen species (ROS) are under scrutiny as a participant in the pathophysiology of myelodysplastic syndrome (MDS) and the progression of MDS to acute myeloid leukemia (AML). Measurement of intracellular ROS (iROS) is particularly important since iROS is a direct indicator of cellular health and integrity.Methods: We developed a technique to measure standardize iROS (siROS) level in lymphocytes and bone marrow (BM) CD34+ hematopoietic progenitors using the fluorescent probe dichlorofluorescein (DCF). We then quantified the siROS in 38 consecutive BM specimens from 27 MDS patients over the course of 10 months. Disease outcome of these patients were also assessed.Results: High serum ferritin, high blast count and poor IPSS were associated with inferior survival and AML progression in this cohort. High blast MDS patients had lower siROS in their BM CD34+ cells than those of low blast patients, consistent with increased reliance on glycolysis and enhanced ROS defense in high blast MDS. We also observed narrower siROS distribution in the BM CD34+ cells of high blast patients, suggesting that loss of heterogeneity in ROS content accompanies the clonal evolution of MDS. Furthermore, we observed a strong correlation between CD34+ cells siROS and serum ferritin level in high blast patients. In one case, iron chelation therapy (ICT) resulted in parallel decreases in serum ferritin and CD34+ cells siROS.Conclusion: Our findings established the siROS profile in early hematopoietic cells of MDS patients and its relationship with blast count and iron overload.


Asunto(s)
Crisis Blástica/metabolismo , Células Madre Hematopoyéticas/metabolismo , Sobrecarga de Hierro/metabolismo , Leucemia Mieloide Aguda/metabolismo , Síndromes Mielodisplásicos/metabolismo , Anciano , Anciano de 80 o más Años , Crisis Blástica/etiología , Crisis Blástica/patología , Crisis Blástica/terapia , Femenino , Células Madre Hematopoyéticas/patología , Humanos , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/terapia , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/complicaciones , Síndromes Mielodisplásicos/patología , Síndromes Mielodisplásicos/terapia
3.
Hematol Oncol Clin North Am ; 34(2): 441-448, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32089221

RESUMEN

BCL-2 is an antiapoptotic protein that plays a critical role acute and chronic leukemias. Venetoclax is an orally selective BCL-2 inhibitor and BH3 mimetic approved in chronic lymphocytic leukemia and in combination with low dose cytarabine or hypomethylating agent in acute myeloid leukemia for the treatment of patients unfit for intensive chemotherapy. This article reviews the biology of BCL-2, focusing on its relationship to the myeloid microenvironment, and discusses the rationale for BCL-2 inhibition in myelodysplastic syndrome (MDS). Clinical trials testing venetoclax in MDS patients are under way. Potential biomarkers for clinical response to BCL-2 inhibition are discussed. Therapeutic opportunities for venetoclax in the therapeutic landscape of MDS are explored.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Síndromes Mielodisplásicos/tratamiento farmacológico , Sulfonamidas/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Estudios Clínicos como Asunto , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Evaluación Preclínica de Medicamentos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/etiología , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/etiología , Síndromes Mielodisplásicos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfonamidas/farmacología , Resultado del Tratamiento
4.
Clin Exp Med ; 20(1): 1-9, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31712933

RESUMEN

Iron overload remains a concern in myelodysplastic syndrome (MDS) patients especially those requiring recurrent blood transfusions. Whether iron chelating therapy (ICT) is beneficial to the long-term survival of myelodysplastic syndrome is still a controversial issue. Therefore, we conducted a systematic review and meta-analysis to clarify the relationship between ICT and long-term survival in patients with MDS. A total of 14 studies involving 7242 participants were identified; the outcomes revealed that for patients with MDS, ICT resulted in a lower risk of mortality compared to those with no ICT (HR 0.57; 95% CI 0.44-0.70; P < 0.001); what is more, ICT led to a lower risk of leukemia transformation (HR 0.70; 95% CI 0.52-0.93; P = 0.016). Results of subgroup analyses based on adequate ICT or any ICT, low/int-1 IPSS or unclassified IPSS and study types indicated that the ICT had a beneficial role in all these groups of patients.


Asunto(s)
Terapia por Quelación/métodos , Sobrecarga de Hierro/tratamiento farmacológico , Síndromes Mielodisplásicos/complicaciones , Humanos , Hierro , Sobrecarga de Hierro/etiología , Síndromes Mielodisplásicos/metabolismo , Análisis de Supervivencia , Resultado del Tratamiento
5.
Hematology ; 25(1): 1-10, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31838956

RESUMEN

Objectives: Background/aims: We aim to explore low-risk MDS patients' ESA response and the difference between iron-overloaded (IO) group and the control group in the expression of SOCS1, STAT5 and BCL2L1 which play a key role to EPO-STAT5 signal pathway.Methods: 56 low-risk MDS patients were divided into experimental group, IO patients; control group, non-IO patients. Among experimental group, 28 IO patients were treated with iron chelation therapy (ICT). SOCS1, phosphorylated STAT5 (p-STAT5) and BCL2L1 protein concentration in bone marrow supernatant have been analyzed by ELISA, STAT5a+b protein concentration in bone marrow mononuclear cells (BMMC) have been analyzed by Western blot, and mRNA expression of them have been detected in BMMC by RQ-PCR. The percentage of CD71+ cells in BMMC, apoptotic rate of CD71+ cells and ROS expression in CD71+ cells were detected by Flow cytometry.Results: Compared with the control group, the sEPO concentration, the efficacy of ESA and the expression of SOCS1, apoptotic rates of CD71+ cells and ROS expression in CD71+ cells in IO group were increased, the expression of STAT5 and BCL2L1 was reduced. Interestingly, after receiving ICT, some patients with EPO resistance have responded again to ESA treatment, with the decrease of the expression of SOCS1, apoptotic rates of CD71+ cells, ROS expression in CD71+ cells and the increase of the expression of STAT5 and BCL2L1.Conclusion: Iron overload can increase EPO resistance and the expression of SOCS1, inhibit the expression of STAT5 and BCL2L1. ICT could allivation of EPO resistance.


Asunto(s)
Eritropoyetina/metabolismo , Hematínicos/uso terapéutico , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Factor de Transcripción STAT5/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Terapia por Quelación , Femenino , Humanos , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/metabolismo , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/complicaciones , Síndromes Mielodisplásicos/metabolismo , Transducción de Señal/efectos de los fármacos
6.
Aging (Albany NY) ; 11(21): 9626-9642, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31727865

RESUMEN

Bone marrow stromal cells from patients with myelodysplastic syndrome (MDS) display a senescence phenotype, but the underlying mechanism has not been elucidated. Pro-inflammatory signaling within the malignant clone and the bone marrow microenvironment has been identified as a key pathogenetic driver of MDS. Our study revealed that S100A9 is highly-expressed in lower-risk MDS. Moreover, normal primary mesenchymal stromal cells (MSCs) and the human stromal cell line HS-27a co-cultured with lower-risk MDS bone marrow mononuclear cells acquired a senescence phenotype. Exogenous supplemented S100A9 also induced cellular senescence in MSCs and HS-27a cells. Importantly, Toll-like receptor 4 (TLR4) inhibition or knockdown attenuated the cellular senescence induced by S100A9. Furthermore, we showed that S100A9 induces NLRP3 inflammasome formation, and IL-1ß secretion; findings in samples from MDS patients further confirmed these thoughts. Moreover, ROS and IL-1ß inhibition suppressed the cellular senescence induced by S100A9, whereas NLRP3 overexpression and exogenous IL-1ß supplementation induces cellular senescence. Our study demonstrated that S100A9 promotes cellular senescence of bone marrow stromal cells via TLR4, NLRP3 inflammasome formation, and IL-1ß secretion for its effects. Our findings deepen the understanding of the molecular mechanisms involved in MDS reprogramming of MSCs and indicated the essential role of S100A9 in tumor-environment interactions in bone marrow.


Asunto(s)
Calgranulina B/metabolismo , Senescencia Celular/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Línea Celular , Células Cultivadas , Reprogramación Celular/fisiología , Femenino , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/etiología , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Nicho de Células Madre/fisiología , Receptor Toll-Like 4/metabolismo , Regulación hacia Arriba , Adulto Joven
7.
Rinsho Ketsueki ; 59(10): 2042-2049, 2018.
Artículo en Japonés | MEDLINE | ID: mdl-30305507

RESUMEN

Myelodysplastic syndromes (MDS) are clonal hematopoietic disorders characterized by ineffective hematopoiesis in bone marrow and cytopenias in peripheral blood. In patients with MDS, iron overload is frequent due to red blood cell transfusions and ineffective erythropoiesis. Dysplastic erythroblasts in MDS secrete humoral factors such as erythroferrone, which suppress hepatic expression of hepcidin. Hepcidin is the key regulator of systemic iron homeostasis, and suppression of hepcidin expression leads to an increase in iron absorption from the intestines, exacerbating systemic iron overload. Patients with MDS with ring sideroblasts (MDS-RS) are prone to iron overload, with most harboring splicing factor 3B subunit 1 (SF3B1) mutations in hematopoietic cells. SF3B1 mutations may induce ring sideroblasts by downregulating ATP binding cassette subfamily B member 7, which exports iron-sulfur clusters from the mitochondria to the cytoplasm. Iron overload in MDS causes hepatic dysfunction, diabetes, cardiac failure, and atherosclerosis, whereas excess iron may suppress normal hematopoiesis. Though randomized control studies are lacking, results from retrospective and cohort studies indicate that iron chelation therapy is appropriate for lower-risk MSD patients with transfusion-related iron overload, although it is not recommended for higher-risk MSD patients with short life expectancy.


Asunto(s)
Terapia por Quelación , Sobrecarga de Hierro/complicaciones , Hierro/metabolismo , Síndromes Mielodisplásicos/complicaciones , Hematopoyesis , Hepcidinas , Humanos , Síndromes Mielodisplásicos/metabolismo , Estudios Retrospectivos
8.
Leuk Res ; 74: 21-41, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30286330

RESUMEN

In 2008 the first evidence-based Canadian consensus guideline addressing the diagnosis, monitoring and management of transfusional iron overload in patients with myelodysplastic syndromes (MDS) was published. The Canadian Consortium on MDS, comprised of hematologists from across Canada with a clinical and academic interest in MDS, reconvened to update these guidelines. A literature search was updated in 2017; topics reviewed include mechanisms of iron overload induced cellular damage, evidence for clinical endpoints impacted by iron overload including organ dysfunction, infections, marrow failure, overall survival, acute myeloid leukemia progression, and endpoints around hematopoietic stem-cell transplant. Evidence for an impact of iron reduction on the same endpoints is discussed, guidelines are updated, and areas identified where evidence is suboptimal. The guidelines address common questions around the diagnosis, workup and management of iron overload in clinical practice, and take the approach of who, when, why and how to treat iron overload in MDS. Practical recommendations for treatment and monitoring are made. Evidence levels and grading of recommendations are provided for all clinical endpoints examined.


Asunto(s)
Sobrecarga de Hierro , Síndromes Mielodisplásicos , Canadá , Femenino , Humanos , Sobrecarga de Hierro/diagnóstico , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Sobrecarga de Hierro/terapia , Masculino , Estudios Multicéntricos como Asunto , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Síndromes Mielodisplásicos/terapia , Guías de Práctica Clínica como Asunto
9.
Blood Rev ; 32(6): 473-479, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29699840

RESUMEN

Excess iron can be extremely toxic for the body and may cause organ damage in the absence of iron chelation therapy. Preclinical studies on the role of free iron on bone marrow function have shown that iron toxicity leads to the accumulation of reactive oxygen species, affects the expression of genes coding for proteins that regulate hematopoiesis, and disrupts hematopoiesis. These effects could be partially attenuated by iron-chelation treatment with deferasirox, suggesting iron toxicity may have a negative impact on the hematopoietic microenvironment. Iron toxicity is of concern in transfusion-dependent patients. Importantly, iron chelation with deferasirox can cause the loss of transfusion dependency and may induce hematological responses, although the mechanisms through which deferasirox exerts this action are currently unknown. This review will focus on the possible mechanisms of toxicity of free iron at the bone marrow level and in the bone marrow microenvironment.


Asunto(s)
Médula Ósea/metabolismo , Susceptibilidad a Enfermedades , Hierro/metabolismo , Anemia Aplásica/complicaciones , Anemia Aplásica/etiología , Anemia Aplásica/metabolismo , Anemia Aplásica/terapia , Animales , Células de la Médula Ósea/metabolismo , Microambiente Celular , Células Madre Hematopoyéticas/metabolismo , Humanos , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/metabolismo , Síndromes Mielodisplásicos/complicaciones , Síndromes Mielodisplásicos/etiología , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/terapia , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/etiología , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/terapia
10.
Cell Rep ; 20(3): 572-585, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28723562

RESUMEN

Myelodysplastic syndromes and chronic myelomonocytic leukemia are blood disorders characterized by ineffective hematopoiesis and progressive marrow failure that can transform into acute leukemia. The DNA methyltransferase inhibitor 5-azacytidine (AZA) is the most effective pharmacological option, but only ∼50% of patients respond. A response only manifests after many months of treatment and is transient. The reasons underlying AZA resistance are unknown, and few alternatives exist for non-responders. Here, we show that AZA responders have more hematopoietic progenitor cells (HPCs) in the cell cycle. Non-responder HPC quiescence is mediated by integrin α5 (ITGA5) signaling and their hematopoietic potential improved by combining AZA with an ITGA5 inhibitor. AZA response is associated with the induction of an inflammatory response in HPCs in vivo. By molecular bar coding and tracking individual clones, we found that, although AZA alters the sub-clonal contribution to different lineages, founder clones are not eliminated and continue to drive hematopoiesis even in complete responders.


Asunto(s)
Azacitidina/administración & dosificación , Resistencia a Medicamentos , Genómica , Síndromes Mielodisplásicos , Anciano , Anciano de 80 o más Años , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/genética , Femenino , Humanos , Cadenas alfa de Integrinas/genética , Cadenas alfa de Integrinas/metabolismo , Persona de Mediana Edad , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo
11.
Expert Rev Hematol ; 10(1): 53-64, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27923273

RESUMEN

INTRODUCTION: Patients with myelodysplastic syndromes (MDS) differ from those with other transfusion-dependent conditions (eg, thalassemia) as they are typically older, have comorbid conditions, and a generally shorter life expectancy. The underlying disease process in MDS and frequent use of red blood cell transfusions lead to iron accumulation and ultimately organ damage. Whether iron-reducing interventions such as chelation therapy can improve outcomes in this population is currently under investigation. Areas covered: We reviewed published English-language articles from PubMed on the topic of iron overload (IO) in MDS, and the use of iron chelation therapies (ICTs) to alleviate iron burden. Expert commentary: Data on IO-associated complications in MDS are derived largely from retrospective studies and there are limited data to guide clinicians on major treatment decisions. Although effective and well-tolerated oral ICTs are available, and general recommendations may be made regarding usage in MDS, guidance is not yet based on prospective data. The clinical endpoints and assessments for MDS may differ substantively from those used in patients with thalassemia, as an older population may have competing causes for morbidity. We expect that emergent data from clinical trials currently underway will define more appropriate endpoints/assessments for the MDS population in clinical trials.


Asunto(s)
Terapia por Quelación , Toma de Decisiones Clínicas , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/terapia , Síndromes Mielodisplásicos/complicaciones , Reacción a la Transfusión , Biopsia , Árboles de Decisión , Humanos , Hierro/metabolismo , Quelantes del Hierro/administración & dosificación , Quelantes del Hierro/efectos adversos , Sobrecarga de Hierro/diagnóstico , Sobrecarga de Hierro/metabolismo , Hígado/metabolismo , Hígado/patología , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/mortalidad , Síndromes Mielodisplásicos/terapia , Tiempo de Tratamiento , Resultado del Tratamiento
12.
J Ethnopharmacol ; 196: 253-260, 2017 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-27988397

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Hedyotis diffusa is an ethno-medicine used for anti-cancer treatment in the clinic of traditional Chinese medicine (TCM). The total coumarins of Hedyotis diffusa (TCHD) was a selected extract with observed antiproliferative activity, which has not been tested in treatment of myelodysplastic syndromes (MDS) or acute myeloid leukemia (AML). AIM OF THE STUDY: This study aimed to evaluate the apoptosis-inducing effect of TCHD on human MDS cell line (SKM-1) and explore its action mechanism in association with caspase family and PI3K/Akt signaling pathway. MATERIALS AND METHODS: The chemical constituents and total coumarins content of TCHD were determined by High Performance Liquid Chromatography-tandem mass spectrometry (HPLC-MS/MS) and UV-vis spectrophotometry, respectively. MTT assay, Hoechst 33258 staining, and Annexin V-FITC/PI double labeling were applied to evaluate TCHD's efficacy on SKM-1 cells. Western blot analysis was also used to clarify the action mechanism of TCHD on protein expression level. RESULTS: Two compounds, p-coumaric acid and E-6-O-p-coumaroyl scandoside methyl ester, were identified in TCHD, and its total coumarins content reached 87.4%. By MTT assay, apoptosis-inducing effect of TCHD on SKM-1 cells was found in a dose-dependent manner after 24-48h treatment, with IC50 values of 104.48µg/ml and 100.66µg/ml, respectively. Morphological and flow cytometry observation also confirmed such effect of TCHD. Western blot analysis clarified its action mechanism associating with the activation of caspases and inhibition of PI3K/Akt pathway proteins. CONCLUSIONS: This is the first report regarding the apoptosis-inducing efficacy and mechanism of TCHD on SKM-1 cells, providing a promising candidate of TCM for MDS and AML therapy with fewer side effects.


Asunto(s)
Cumarinas/farmacología , Hedyotis , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Síndromes Mielodisplásicos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
13.
BMJ Support Palliat Care ; 6(1): 80-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25204541

RESUMEN

OBJECTIVE: The primary objective of this study was to evaluate the health-related quality of life (HRQOL) in lower-risk, transfusion-dependent patients with myelodysplastic syndromes (MDS) treated with deferasirox. A secondary objective was to investigate the relationship between HRQOL, serum ferritin levels and transfusion dependency. PATIENTS AND METHODS: This was a prospective multicentre study enrolling 159 patients, of whom 152 received at least one dose of deferasirox. HRQOL was assessed with the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire-Core 30 (EORTC QLQ-C30) at baseline and then at 3, 6, 9 and 12 months. Primary analysis was performed estimating mean HRQOL scores over time by a linear mixed model on selected scales. RESULTS: The median age of treated patients was 72 years (range 24-87 years). No statistically significant changes over time were found in mean scores for global health status/quality of life (p=0.564), physical functioning (p=0.409) and fatigue (p=0.471) scales. Also, no significant changes were found for constipation (p=0.292), diarrhoea (p=0.815) and nausea and vomiting (p=0.643). Serum ferritin levels were not associated with HRQOL outcomes. A higher patient-reported baseline pain severity was an independent predictive factor of an earlier achievement of transfusion independence with a HR of 1.032 (99% CI 1.004 to 1.060; p=0.003). CONCLUSIONS: HRQOL of transfusion-dependent patients with MDS receiving deferasirox therapy remains stable over time. HRQOL assessment might also provide important predictive information on treatment outcomes. TRIAL REGISTRATION NUMBER: NCT00469560.


Asunto(s)
Benzoatos/uso terapéutico , Quelantes del Hierro/uso terapéutico , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/psicología , Calidad de Vida , Reacción a la Transfusión , Triazoles/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Benzoatos/administración & dosificación , Deferasirox , Femenino , Humanos , Quelantes del Hierro/administración & dosificación , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/metabolismo , Estudios Prospectivos , Resultado del Tratamiento , Triazoles/administración & dosificación , Adulto Joven
14.
Oncotarget ; 6(32): 32494-508, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26378812

RESUMEN

We have previously shown that a specific promyelocytic leukemia-retinoic acid receptor alpha (PML-RARA) DNA vaccine combined with all-trans retinoic acid (ATRA) increases the number of long term survivors with enhanced immune responses in a mouse model of acute promyelocytic leukemia (APL). This study reports the efficacy of a non-specific DNA vaccine, pVAX14Flipper (pVAX14), in both APL and high risk myelodysplastic syndrome (HR-MDS) models. PVAX14 is comprised of novel immunogenic DNA sequences inserted into the pVAX1 therapeutic plasmid. APL mice treated with pVAX14 combined with ATRA had increased survival comparable to that obtained with a specific PML-RARA vaccine. Moreover, the survival advantage correlated with decreased PML-RARA transcript levels and increase in anti-RARA antibody production. In HR-MDS mice, pVAX14 significantly improved survival and reduced biomarkers of leukemic transformation such as phosphorylated mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK) 1. In both preclinical models, pVAX14 vaccine significantly increased interferon gamma (IFNγ) production, memory T-cells (memT), reduced the number of colony forming units (CFU) and increased expression of the adapter molecule signalling to NF-κB, MyD88. These results demonstrate the adjuvant properties of pVAX14 providing thus new approaches to improve clinical outcome in two different models of myeloid malignancies, which may have potential for a broader applicability in other cancers.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacunas contra el Cáncer/farmacología , Leucemia Promielocítica Aguda/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Neoplasias Experimentales/tratamiento farmacológico , Tretinoina/farmacología , Vacunas de ADN/farmacología , Animales , Anticuerpos/sangre , Secuencia de Bases , Vacunas contra el Cáncer/inmunología , Regulación Neoplásica de la Expresión Génica , Genes ras , Memoria Inmunológica/efectos de los fármacos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/inmunología , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones Transgénicos , Datos de Secuencia Molecular , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/inmunología , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Vacunación , Vacunas de ADN/inmunología
15.
Acta Haematol ; 134(4): 233-42, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26138856

RESUMEN

Iron overload in transfusion-dependent patients with rare anemias can be managed with chelation therapy. This study evaluated deferasirox efficacy and safety in patients with myelodysplastic syndromes (MDS), aplastic anemia (AA) or other rare anemias. A 1-year, open-label, multicenter, single-arm, phase II trial was performed with deferasirox (10­40 mg/kg/day, based on transfusion frequency and therapeutic goals), including an optional 1-year extension. The primary end point was a change in liver iron concentration (LIC) after 1 year. Secondary end points included changes in efficacy and safety parameters (including ophthalmologic assessments) overall as well as in a Japanese subpopulation. Overall, 102 patients (42 with MDS, 29 with AA and 31 with other rare anemias) were enrolled; 57 continued into the extension. Mean absolute change in LIC was ­10.9 mg Fe/g dry weight (d.w.) after 1 year (baseline: 24.5 mg Fe/g d.w.) and ­13.5 mg Fe/g d.w. after 2 years. The most common drug-related adverse event was increased serum creatinine (23.5%), predominantly in MDS patients. Four patients had suspected drug-related ophthalmologic abnormalities. Outcomes in Japanese patients were generally consistent with the overall population. Results confirm deferasirox efficacy in patients with rare anemias, including a Japanese subpopulation. The safety profile was consistent with previous studies and ophthalmologic parameters generally agreed with baseline values (EUDRACT 2006-003337-32).


Asunto(s)
Anemia Aplásica/tratamiento farmacológico , Benzoatos/administración & dosificación , Sobrecarga de Hierro/tratamiento farmacológico , Hígado/metabolismo , Síndromes Mielodisplásicos/tratamiento farmacológico , Triazoles/administración & dosificación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anemia Aplásica/metabolismo , Anemia Aplásica/patología , Benzoatos/efectos adversos , Niño , Preescolar , Deferasirox , Humanos , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Hígado/patología , Persona de Mediana Edad , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Triazoles/efectos adversos
16.
Ann Hematol ; 94(5): 771-7, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25743685

RESUMEN

Iron chelation therapy can improve hematopoiesis in myelodysplastic syndromes. Only few studies showed hematologic improvement with deferoxamine, and the erythroid responses were correlated with good compliance to long-term treatment. Indeed, single-case reports and data from clinical trials testing the efficacy of deferasirox reported hematologic improvements with varying rates of response in different lineages. Overall, about 760 myelodysplastic syndrome (MDS) patients with iron overload receiving deferasirox were included in six different studies, and an increase in hemoglobin level was reported to range from 6 to 44.5%, an increase in platelet count from 13 to 61%, and in neutrophil count from 3 to 76%. In all the published studies, hematologic improvements were not related to serum ferritin or to non-total binding iron changes; indeed, other pathways were indicated as possible pathogenetic mechanisms, such as decreased NF-kB activity, modulation of mTOR signalling, and reduced reactive oxygen species. The aims of this review are to provide all available information relating clinical and hematologic changes after chelation therapy and to discuss potential mechanisms involved in such responses.


Asunto(s)
Plaquetas/metabolismo , Terapia por Quelación , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/metabolismo , Leucocitos/metabolismo , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/metabolismo , Benzoatos/uso terapéutico , Deferasirox , Hemoglobinas/metabolismo , Humanos , Quelantes del Hierro/uso terapéutico , Triazoles/uso terapéutico
17.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 22(4): 1027-32, 2014 Aug.
Artículo en Chino | MEDLINE | ID: mdl-25130822

RESUMEN

This study was aimed to investigate the changes of erythropoietin (EPO), hemoglobin(Hb) and recombinant EPO (rEPO) levels in MDS patients receiving iron chelation therapy, and to explore the relationship between EPO and serum ferritin(SF). A total of 172 MDS patients and 30 healthy controls were studied. The levels of SF, EPO, serum iron (SI), total iron binding capacity (TIBC), C-reaction protein (CRP) and Hb were measured respectively, the level of SF was adjusted according to the changes of CRP. Among them, there were 34 cases of low-risk (SF>1 000 mg/L) receiving deferoxamine therapy, whose changes of SF, EPO, SI, TIBC, Hb levels were detected and compared before and after treatment. Besides, the difference in the incidence of EPO resistance in iron overload group and non-iron overload group was assessed before and after therapy, and 58 cases of low-risk and EPO<1 000 U/L MDS patients were given rEPO therapy. The results showed that the level of EPO in non-iron overload group was higher than that in the normal control group (997.44 ± 473.48 vs 467.27 ± 238.49, P < 0.05). Obviously, the level of EPO in iron overload group was higher than that in non-iron overload group and control group (3257.59 ± 697.19 vs 997.44 ± 473.48, P = 0.012, 3257.59 ± 697.19 vs 467.27 ± 238.49, P = 0.002). Otherwise, the incidence of EPO resistance in iron overload group was higher than that in non-iron overload group (18/35 vs 2/23, P = 0.001), and the level of EPO and SF was positively related to each other in iron overload group (r = 0.310,P = 0.036). After receiving iron chelation therapy, the levels of SF, SI, TIBC and EPO in iron overload group were significantly lower than that before therapy (3942.38 ± 641.82 vs 2266.35 ± 367.31, P = 0.028;48.61 ± 10.65 vs 28.52 ± 12.61, P = 0.034;59.84 ± 12.62 vs 33.76 ± 15.43, P = 0.045;3808.01 ± 750.22 vs 1954.78 ± 473.18, P = 0.042). Moreover, the level of Hb increased (35 ± 18 vs 57 ± 21, P = 0.046) and the EPO resistance in some patients was decreased. It is concluded that iron chelation therapy can improve the efficacy of EPO to alleviate EPO resistance in patients wtih anemic MDS, decrease the pathological level of EPO, enhance Hb levels and reduce the dependency on blood transfusion.


Asunto(s)
Terapia por Quelación , Eritropoyetina/sangre , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/metabolismo , Adulto , Anciano , Proteína C-Reactiva/metabolismo , Estudios de Casos y Controles , Femenino , Ferritinas/sangre , Hemoglobinas/metabolismo , Humanos , Hierro/metabolismo , Sobrecarga de Hierro , Masculino , Persona de Mediana Edad , Proteínas Recombinantes/uso terapéutico
18.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 34(2): 174-8, 2014 Feb.
Artículo en Chino | MEDLINE | ID: mdl-24672941

RESUMEN

OBJECTIVE: To study the effect of Qinghuang Powder (QHP) combined Chinese herbs for Pi-strengthening and Shen-reinforcing (CHPSSR) on hypoxia-inducible factor 1alpha (HIF-1alpha) in bone marrow mononuclear cells of myelodysplastic syndrome (MDS) patients. METHODS: Changes of HIF-1alpha in bone marrow mononuclear cells of MDS patients were detected in 25 MDS patients treated by QHP combined CHPSSR using flow cytometry. Meanwhile, 13 healthy subjects were recruited as the control group. Changes HIF-1alpha levels in various serial bone marrow mononuclear cells were detected. RESULTS: (1) Among the 25 patients in the treatment group, there were 19 patients effective and 6 patients ineffective, with the total effective rate being 76%. (2) Compared with before treatment, levels of peripheral blood WBC, Hb, PLT, and ANC significantly increased in the treatment group after treatment, showing statistical difference (P < 0.05, P < 0.01). (3) Compared with before treatment, the HIF-1alpha mean fluorescence intensity was enhanced in bone marrow lymphocytes, monocytes, granulocytes, and nucleated red blood cells of the treatment group after treatment (P < 0.05, P < 0.01). Compared with the control group, the HIF-1alpha mean fluorescence intensity was weakened in bone marrow lymphocytes, monocytes, and nucleated red blood cells of the treatment group before treatment; while it was obviously enhanced in granulocytes (P < 0.01). But after treatment the HIF-1alpha mean fluorescence intensity increased more in the granulocytes of the treatment group than in those of the control group (P < 0.01), but there was no statistical difference in bone marrow lymphocytes, monocytes, or nucleated red blood cells. CONCLUSION: QHP combined CHPSSR could improve HIF-1alpha levels in bone marrow lymphocytes, monocytes, granulocytes, and nucleated red blood cells of MDS patients, thus improving Hb levels of MDS patients, and improving their anemia and correlated symptoms.


Asunto(s)
Arsenicales/uso terapéutico , Células de la Médula Ósea/metabolismo , Medicamentos Herbarios Chinos/uso terapéutico , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Monocitos/metabolismo , Síndromes Mielodisplásicos/metabolismo , Adolescente , Adulto , Anciano , Médula Ósea , Células de la Médula Ósea/efectos de los fármacos , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Síndromes Mielodisplásicos/tratamiento farmacológico , Fitoterapia , Adulto Joven
19.
Expert Rev Anticancer Ther ; 14(7): 817-29, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24641787

RESUMEN

Transfusion dependent low risk myelodysplastic syndromes (MDS) patients, eventually develop iron overload. Iron toxicity, via oxidative stress, can damage cellular components and impact organ function. In thalassemia major patients, iron chelation therapy lowered iron levels with recovery of cardiac and liver functions and significant improvement in survival. Several noncontrolled studies show inferior survival in MDS patients with iron overload, including an increase in transplant-related mortality and infection risk while iron chelation appears to improve survival in both lower risk MDS patients and in stem cell transplant settings. Collated data are presented on the pathophysiological impact of iron overload; measuring techniques and chelating agents' therapy positive impact on hematological status and overall survival are discussed. Although suggested by retrospective analyses, the lack of clear prospective data of the beneficial effects of iron chelation on morbidity and survival, the role of iron chelation therapy in MDS patients remains controversial.


Asunto(s)
Terapia por Quelación , Quelantes del Hierro/uso terapéutico , Síndromes Mielodisplásicos/tratamiento farmacológico , Administración Oral , Benzoatos/farmacología , Transfusión Sanguínea , Deferasirox , Deferiprona , Deferoxamina/uso terapéutico , Ferritinas/sangre , Humanos , Hierro/análisis , Hierro/metabolismo , Quelantes del Hierro/administración & dosificación , Quelantes del Hierro/efectos adversos , Sobrecarga de Hierro/fisiopatología , Leucemia Mieloide Aguda/etiología , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/mortalidad , Piridonas/uso terapéutico , Triazoles/farmacología
20.
Expert Opin Investig Drugs ; 23(2): 255-63, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24410313

RESUMEN

INTRODUCTION: Clofarabine is a second-generation purine nucleoside analog approved in 2004 for the treatment of pediatric patients with relapsed or refractory acute lymphocytic leukemia (ALL) following failure of at least two prior regimens. Clofarabine is a hybrid of fludarabine and cladribine, designed to overcome the pharmacologic limitations associated with its predecessors, while retaining their beneficial properties. In addition to providing a valuable treatment option for pediatric patients with ALL, clofarabine alone and in combination with cytarabine (Ara-C) has demonstrated substantial activity against myelodysplastic syndrome (MDS), thus rendering this agent a potential therapeutic option for MDS. AREAS COVERED: This review focuses on the pharmacology and clinical activity of clofarabine in MDS, as well as its emerging role in the treatment of MDS. Publications in English were selected from the MEDLINE (PubMed) database, as well articles of interest from bibliographies and abstracts based on the publication of meeting materials. EXPERT OPINION: DNA-methyltransferase inhibitors are the mainstay of therapy for many patients with MDS who require treatment. Although these agents are very well tolerated and represent a significant advancement in the treatment of MDS by improving transfusion requirements and prolonging survival in various subgroups of patients, response rates are modest and the duration of response is short. In addition to providing a valuable treatment option for pediatric ALL patients, clofarabine has substantial activity against MDS and is well tolerated by elderly patients, thus rendering it a potential therapeutic option.


Asunto(s)
Nucleótidos de Adenina/uso terapéutico , Antimetabolitos Antineoplásicos/uso terapéutico , Arabinonucleósidos/uso terapéutico , Síndromes Mielodisplásicos/tratamiento farmacológico , Nucleótidos de Adenina/farmacocinética , Nucleótidos de Adenina/farmacología , Animales , Antimetabolitos Antineoplásicos/farmacocinética , Antimetabolitos Antineoplásicos/farmacología , Arabinonucleósidos/farmacocinética , Arabinonucleósidos/farmacología , Clofarabina , Citarabina/uso terapéutico , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Humanos , Síndromes Mielodisplásicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA