Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Blood Rev ; 64: 101165, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38182489

RESUMEN

α-Thalassemia is an inherited blood disorder characterized by decreased synthesis of α-globin chains that results in an imbalance of α and ß globin and thus varying degrees of ineffective erythropoiesis, decreased red blood cell (RBC) survival, chronic hemolytic anemia, and subsequent comorbidities. Clinical presentation varies depending on the genotype, ranging from a silent or mild carrier state to severe, transfusion-dependent or lethal disease. Management of patients with α-thalassemia is primarily supportive, addressing either symptoms (eg, RBC transfusions for anemia), complications of the disease, or its transfusion-dependence (eg, chelation therapy for iron overload). Several novel therapies are also in development, including curative gene manipulation techniques and disease modifying agents that target ineffective erythropoiesis and chronic hemolytic anemia. This review of α-thalassemia and its various manifestations provides practical information for clinicians who practice beyond those regions where it is found with high frequency.


Asunto(s)
Enfermedades Hematológicas , Sobrecarga de Hierro , Talasemia alfa , Talasemia beta , Humanos , Talasemia beta/terapia , Talasemia alfa/diagnóstico , Talasemia alfa/genética , Talasemia alfa/terapia , Eritropoyesis , Transfusión de Eritrocitos , Sobrecarga de Hierro/diagnóstico , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/terapia
2.
Expert Rev Hematol ; 14(7): 633-644, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34324404

RESUMEN

INTRODUCTION: In ß-thalassemia, imbalanced globin synthesis causes reduced red blood cell survival and ineffective erythropoiesis. Suppressed hepcidin levels increase ferroportin-mediated iron transport in enterocytes, causing increased iron absorption and potentially iron overload. Low hepcidin also stimulates ferroportin-mediated iron release from macrophages, increasing transferrin saturation (TSAT), potentially forming non-transferrin-bound iron, which can be toxic. Modulating the hepcidin-ferroportin axis is an attractive strategy to improve ineffective erythropoiesis and limit the potential tissue damage resulting from iron overload. There are no oral ß-thalassemia treatments that consistently ameliorate anemia and prevent iron overload. AREAS COVERED: The preclinical and clinical development of vamifeport (VIT-2763), a novel ferroportin inhibitor, was reviewed. PubMed, EMBASE and ClinicalTrials.gov were searched using the search term 'VIT-2763'. EXPERT OPINION: Vamifeport is the first oral ferroportin inhibitor in clinical development. In healthy volunteers, vamifeport had comparable safety to placebo, was well tolerated and rapidly decreased iron levels and reduced TSAT, consistent with observations in preclinical models. Data from ongoing/planned Phase II studies are critical to define its potential in ß-thalassemia and other conditions associated with iron overabsorption and/or ineffective erythropoiesis. If vamifeport potentially increases hemoglobin and reduces iron-related parameters, it could be a suitable treatment for non-transfusion-dependent and transfusion-dependent ß-thalassemia.


Asunto(s)
Sobrecarga de Hierro , Talasemia beta , Proteínas de Transporte de Catión , Eritropoyesis , Hepcidinas/farmacología , Hepcidinas/uso terapéutico , Homeostasis , Humanos , Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Talasemia beta/tratamiento farmacológico
3.
N Engl J Med ; 384(22): 2166, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34077657
5.
N Engl J Med ; 384(3): 252-260, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33283989

RESUMEN

Transfusion-dependent ß-thalassemia (TDT) and sickle cell disease (SCD) are severe monogenic diseases with severe and potentially life-threatening manifestations. BCL11A is a transcription factor that represses γ-globin expression and fetal hemoglobin in erythroid cells. We performed electroporation of CD34+ hematopoietic stem and progenitor cells obtained from healthy donors, with CRISPR-Cas9 targeting the BCL11A erythroid-specific enhancer. Approximately 80% of the alleles at this locus were modified, with no evidence of off-target editing. After undergoing myeloablation, two patients - one with TDT and the other with SCD - received autologous CD34+ cells edited with CRISPR-Cas9 targeting the same BCL11A enhancer. More than a year later, both patients had high levels of allelic editing in bone marrow and blood, increases in fetal hemoglobin that were distributed pancellularly, transfusion independence, and (in the patient with SCD) elimination of vaso-occlusive episodes. (Funded by CRISPR Therapeutics and Vertex Pharmaceuticals; ClinicalTrials.gov numbers, NCT03655678 for CLIMB THAL-111 and NCT03745287 for CLIMB SCD-121.).


Asunto(s)
Anemia de Células Falciformes/terapia , Sistemas CRISPR-Cas , Hemoglobina Fetal/biosíntesis , Edición Génica/métodos , Terapia Genética , Proteínas Represoras/genética , Talasemia beta/terapia , Adulto , Anemia de Células Falciformes/genética , Femenino , Hemoglobina Fetal/genética , Humanos , Proteínas Represoras/metabolismo , Adulto Joven , Talasemia beta/genética
6.
N Engl J Med ; 382(13): 1219-1231, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32212518

RESUMEN

BACKGROUND: Patients with transfusion-dependent ß-thalassemia need regular red-cell transfusions. Luspatercept, a recombinant fusion protein that binds to select transforming growth factor ß superfamily ligands, may enhance erythroid maturation and reduce the transfusion burden (the total number of red-cell units transfused) in such patients. METHODS: In this randomized, double-blind, phase 3 trial, we assigned, in a 2:1 ratio, adults with transfusion-dependent ß-thalassemia to receive best supportive care plus luspatercept (at a dose of 1.00 to 1.25 mg per kilogram of body weight) or placebo for at least 48 weeks. The primary end point was the percentage of patients who had a reduction in the transfusion burden of at least 33% from baseline during weeks 13 through 24 plus a reduction of at least 2 red-cell units over this 12-week interval. Other efficacy end points included reductions in the transfusion burden during any 12-week interval and results of iron studies. RESULTS: A total of 224 patients were assigned to the luspatercept group and 112 to the placebo group. Luspatercept or placebo was administered for a median of approximately 64 weeks in both groups. The percentage of patients who had a reduction in the transfusion burden of at least 33% from baseline during weeks 13 through 24 plus a reduction of at least 2 red-cell units over this 12-week interval was significantly greater in the luspatercept group than in the placebo group (21.4% vs. 4.5%, P<0.001). During any 12-week interval, the percentage of patients who had a reduction in transfusion burden of at least 33% was greater in the luspatercept group than in the placebo group (70.5% vs. 29.5%), as was the percentage of those who had a reduction of at least 50% (40.2% vs. 6.3%). The least-squares mean difference between the groups in serum ferritin levels at week 48 was -348 µg per liter (95% confidence interval, -517 to -179) in favor of luspatercept. Adverse events of transient bone pain, arthralgia, dizziness, hypertension, and hyperuricemia were more common with luspatercept than placebo. CONCLUSIONS: The percentage of patients with transfusion-dependent ß-thalassemia who had a reduction in transfusion burden was significantly greater in the luspatercept group than in the placebo group, and few adverse events led to the discontinuation of treatment. (Funded by Celgene and Acceleron Pharma; BELIEVE ClinicalTrials.gov number, NCT02604433; EudraCT number, 2015-003224-31.).


Asunto(s)
Receptores de Activinas Tipo II/uso terapéutico , Transfusión de Eritrocitos/estadística & datos numéricos , Hematínicos/uso terapéutico , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Talasemia beta/tratamiento farmacológico , Receptores de Activinas Tipo II/efectos adversos , Adolescente , Adulto , Anciano , Método Doble Ciego , Femenino , Ferritinas/sangre , Hematínicos/efectos adversos , Humanos , Fragmentos Fc de Inmunoglobulinas/efectos adversos , Análisis de Intención de Tratar , Análisis de los Mínimos Cuadrados , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Proteínas Recombinantes de Fusión/efectos adversos , Esplenectomía , Adulto Joven , Talasemia beta/genética , Talasemia beta/cirugía , Talasemia beta/terapia
7.
Sci Transl Med ; 11(506)2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31434755

RESUMEN

In ß-thalassemia, accumulated free α-globin forms intracellular precipitates that impair erythroid cell maturation and viability. Protein quality control systems mitigate ß-thalassemia pathophysiology by degrading toxic free α-globin, although the associated mechanisms are poorly understood. We show that loss of the autophagy-activating Unc-51-like kinase 1 (Ulk1) gene in ß-thalassemic mice reduces autophagic clearance of α-globin in red blood cell precursors and exacerbates disease phenotypes, whereas inactivation of the canonical autophagy-related 5 (Atg5) gene has relatively minor effects. Systemic treatment with the mTORC1 inhibitor rapamycin reduces α-globin precipitates and lessens pathologies in ß-thalassemic mice via an ULK1-dependent pathway. Similarly, rapamycin reduces free α-globin accumulation in erythroblasts derived from CD34+ cells of ß-thalassemic individuals. Our findings define a drug-regulatable pathway for ameliorating ß-thalassemia.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Autofagia , Globinas alfa/metabolismo , Talasemia beta/enzimología , Talasemia beta/patología , Animales , Antígenos CD34/metabolismo , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia/metabolismo , Progresión de la Enfermedad , Activación Enzimática/efectos de los fármacos , Eliminación de Gen , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones , Fenotipo , Reticulocitos/efectos de los fármacos , Reticulocitos/metabolismo , Reticulocitos/ultraestructura , Sirolimus/farmacología
8.
Hematology Am Soc Hematol Educ Program ; 2017(1): 278-283, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-29222267

RESUMEN

ß-Thalassemias are characterized by reduced production of ß-globin chain, resulting in α/ß-chain unbalance and precipitation of α-globin-heme complexes and determining ineffective erythropoiesis. Ineffective erythropoiesis, chronic hemolytic anemia, and compensatory hematopoietic expansion are the disease hallmarks, and they are related to the severity of the chain unbalance. Several clinical forms of ß-thalassemia, including the coinheritance of ß-thalassemia with hemoglobin E resulting in hemoglobin E/ß-thalassemia, have been described. Clinically, ß-thalassemias can be classified as transfusion-dependent thalassemia (TDT) and non-transfusion-dependent thalassemia (NTDT) according to the severity of the phenotype, which is caused by a wide spectrum of mutations in a homozygous or compound heterozygous state. Current treatment of TDT consists of regular transfusions that lead to iron overload, requiring iron chelation to prevent iron-related organ toxicity. NTDT patients do not require transfusions or only occasionally require them; however, they develop iron overload as well because of increased intestinal iron absorption caused by chronic anemia. Hematopoietic stem cell allogenic transplant is the only approved cure for ß-thalassemia; however, it is still limited by clinical conditions and the availability of matched donors as well as by potential graft-versus-host disease (GVHD). Gene therapy could avoid the GVHD risk, although hematopoietic stem cells must be genetically modified ex vivo. Epigenetic manipulation and genomic editing are novel experimental approaches. An increased understanding of the pathophysiology that controls the disease process prompted us to explore alternative therapeutic approaches that address the underlying chain unbalance, ineffective erythropoiesis, and iron dysregulation. Molecules, such as JAK2 inhibitors and the activin-receptor ligand trap that target ineffective erythropoiesis, are already in clinical trials with promising results. Other agents aimed to generate iron-restricted erythropoiesis are also under experimental evaluation.


Asunto(s)
Transfusión Sanguínea , Terapia Genética , Quelantes del Hierro/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Talasemia beta/clasificación , Talasemia beta/terapia , Eritropoyesis/efectos de los fármacos , Eritropoyesis/genética , Hemoglobina E/genética , Humanos , Absorción Intestinal , Hierro/metabolismo , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/terapia , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/metabolismo , Mutación , Talasemia beta/sangre , Talasemia beta/genética
11.
Blood Cells Mol Dis ; 57: 23-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26852651

RESUMEN

Efficacy and safety of iron chelation therapy with deferasirox in iron-overloaded non-transfusion-dependent thalassaemia (NTDT) patients were established in the THALASSA study. THETIS, an open-label, single-arm, multicentre, Phase IV study, added to this evidence by investigating earlier dose escalation by baseline liver iron concentration (LIC) (week 4: escalation according to baseline LIC; week 24: adjustment according to LIC response, maximum 30mg/kg/day). The primary efficacy endpoint was absolute change in LIC from baseline to week 52. 134 iron-overloaded non-transfusion-dependent anaemia patients were enrolled and received deferasirox starting at 10mg/kg/day. Mean actual dose±SD over 1year was 14.70±5.48mg/kg/day. At week 52, mean LIC±SD decreased significantly from 15.13±10.72mg Fe/g dw at baseline to 8.46±6.25mg Fe/g dw (absolute change from baseline, -6.68±7.02mg Fe/g dw [95% CI: -7.91, -5.45]; P<0.0001). Most common drug-related adverse events were gastrointestinal: abdominal discomfort, diarrhoea and nausea (n=6 each). There was one death (pneumonia, not considered drug related). With significant and clinically relevant reductions in iron burden alongside a safety profile similar to that in THALASSA, these data support earlier escalation with higher deferasirox doses in iron-overloaded non-transfusion-dependent anaemia patients.


Asunto(s)
Benzoatos/administración & dosificación , Terapia por Quelación/métodos , Quelantes del Hierro/administración & dosificación , Sobrecarga de Hierro/tratamiento farmacológico , Hígado/efectos de los fármacos , Talasemia/tratamiento farmacológico , Triazoles/administración & dosificación , Adolescente , Adulto , Benzoatos/efectos adversos , Transfusión Sanguínea , Niño , Deferasirox , Diarrea/inducido químicamente , Diarrea/diagnóstico , Esquema de Medicación , Cálculo de Dosificación de Drogas , Femenino , Estudios de Seguimiento , Humanos , Hierro/metabolismo , Quelantes del Hierro/efectos adversos , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/patología , Hígado/metabolismo , Hígado/patología , Masculino , Persona de Mediana Edad , Náusea/inducido químicamente , Náusea/diagnóstico , Talasemia/complicaciones , Talasemia/patología , Resultado del Tratamiento , Triazoles/efectos adversos
12.
Crit Rev Oncol Hematol ; 99: 261-71, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26806144

RESUMEN

Iron overload is a potentially life-threatening consequence of multiple red-blood-cell transfusions. Here, we review factors affecting excess iron distribution and its damage to specific tissues, as well as mechanisms of oncogenesis by iron. Although consequences of transfusional iron overload are best described in thalassemia major and related inherited anemias, they are increasingly recognized in acquired conditions, such as myelodysplastic syndromes (MDS). Iron overload in MDS not only impacts on certain tissues, but may affect the clonal evolution of MDS through generation of reactive oxygen species. Iron overload may also influence hematopoietic-stem-cell-transplantation outcomes. Novel MRI methods for assessing body iron have impacted significantly on outcome in inherited anemias by allowing monitoring of iron burden and iron chelation therapy. This approach is increasingly being used in MDS and stem-cell-transplant procedures. Knowledge gained from managing transfusional iron overload in inherited anemias may be translated to general oncology, with potential for improved patient outcomes.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/efectos adversos , Sobrecarga de Hierro/etiología , Hierro/efectos adversos , Síndromes Mielodisplásicos/tratamiento farmacológico , Reacción a la Transfusión , Humanos , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Síndromes Mielodisplásicos/complicaciones
13.
Eur J Intern Med ; 28: 91-6, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26545830

RESUMEN

BACKGROUND: Patients with non-transfusion-dependent thalassemia (NTDT) present with a spectrum of disease severities. Since there are multiple pathophysiologies in such patients, tailoring treatment remains essential. Therefore, one simple, reliable tool would be beneficial to assess disease severity and tailor therapy, particularly for internal medicine specialists who may treat a variety of NTDT patients with a multitude of complications. This would allow for standardization of assessments leading to timely interventions and prevention of complications. METHODS: A working group of NTDT experts was formed to develop a new disease severity scoring system for adult and pediatric patients with NTDT, based on parameters considered to be most pertinent in defining disease severity. RESULTS: 20 parameters were selected for inclusion in the disease severity scoring system. An additional six parameters, largely related to growth and development, were selected specifically for pediatric patients (≤ 16 years of age). Consensus of expert opinion was used to establish the selected methods of assessment for each parameter, based on feasibility and availability of technology, cost containment, and avoidance of patient risk. CONCLUSION: We propose that this new disease severity scoring system for adult and pediatric NTDT patients could be developed into a practical tool for widespread clinical use.


Asunto(s)
Alanina Transaminasa/sangre , Bilirrubina/sangre , Hemoglobinas/metabolismo , Hierro/sangre , Talasemia beta/sangre , Adolescente , Adulto , Arritmias Cardíacas/epidemiología , Enfermedades Óseas Metabólicas/epidemiología , Niño , Colecistectomía , Colecistitis/epidemiología , Colelitiasis/epidemiología , Comorbilidad , Insuficiencia Cardíaca/epidemiología , Hepatitis B/epidemiología , Hepatitis C/epidemiología , Humanos , Hipertensión Pulmonar/epidemiología , Úlcera de la Pierna/epidemiología , Cirrosis Hepática/epidemiología , Osteoporosis/epidemiología , Recuento de Plaquetas , Índice de Severidad de la Enfermedad , Esplenomegalia/epidemiología , Volumen Sistólico , Trombosis/epidemiología , Talasemia beta/epidemiología
14.
Semin Hematol ; 52(4): 261-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26404438

RESUMEN

Anemia is a global public health problem affecting both developing and developed countries at all ages. According to the World Health Organization (WHO), anemia is defined as hemoglobin (Hb) levels <12.0 g/dL in women and <13.0 g/dL in men. However, normal Hb distribution varies not only with sex but also with ethnicity and physiological status. New lower limits of normal Hb values have been proposed, according to ethnicity, gender, and age. Anemia is often multifactorial and is not an independent phenomenon. For the classification and diagnosis the hematologic parameters, the underlying pathological mechanism and patient history should be taken into account. The aging of population, especially in Western countries, causes an increase of anemia in elderly people. In this population, anemia, recently defined by levels of Hb <12 g/dL in both sexes, is mostly of mild degree (10-12 g/dL). Understanding the pathophysiology of anemia in this population is important because it contributes to morbidity and mortality. In one third of the patients, anemia is due to nutritional deficiency, including iron, folate, or vitamin B12 deficiency; moreover, anemia of chronic disease accounts for about another third of the cases. However, in one third of patients anemia cannot be explained by an underlying disease or by a specific pathological process, and for this reason it is defined "unexplained anemia". Unexplained anemia might be due to the progressive resistance of bone marrow erythroid progenitors to erythropoietin, and a chronic subclinical pro-inflammatory state.


Asunto(s)
Anemia , Envejecimiento , Anemia/diagnóstico , Anemia/tratamiento farmacológico , Anemia/epidemiología , Biomarcadores/sangre , Senescencia Celular , Eritropoyetina/uso terapéutico , Humanos , Prevalencia , Células Madre/citología
15.
Blood ; 125(25): 3868-77, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-25934475

RESUMEN

Deferasirox (DFX) monotherapy is effective for reducing myocardial and liver iron concentrations (LIC), although some patients may require intensive chelation for a limited duration. HYPERION, an open-label single-arm prospective phase 2 study, evaluated combination DFX-deferoxamine (DFO) in patients with severe transfusional myocardial siderosis (myocardial [m] T2* 5-<10 ms; left ventricular ejection fraction [LVEF] ≥56%) followed by optional switch to DFX monotherapy when achieving mT2* >10 ms. Mean dose was 30.5 mg/kg per day DFX and 36.3 mg/kg per day DFO on a 5-day regimen. Geometric mean mT2* ratios (Gmeanmonth12/24/Gmeanbaseline) were 1.09 and 1.30, respectively, increasing from 7.2 ms at baseline (n = 60) to 7.7 ms at 12 (n = 52) and 9.5 ms at 24 months (n = 36). Patients (17 of 60; 28.3%) achieved mT2* ≥10 ms and ≥10% increase from baseline at month 24; 15 switched to monotherapy during the study based on favorable mT2*. LIC decreased substantially from a baseline of 33.4 to 12.8 mg Fe/g dry weight at month 24 (-52%). LVEF remained stable with no new arrhythmias/cardiac failure. Five patients discontinued with mT2* <5 ms and 1 died (suspected central nervous system infection). Safety was consistent with established monotherapies. Results show clinically meaningful improvements in mT2* in about one-third of patients remaining on treatment at month 24, alongside rapid decreases in LIC in this heavily iron-overloaded, difficult-to-treat population. Combination therapy may be useful when rapid LIC reduction is required, regardless of myocardial iron overload. This trial was registered at www.clinicaltrials.gov as #NCT01254227.


Asunto(s)
Benzoatos/administración & dosificación , Deferoxamina/administración & dosificación , Quelantes del Hierro/administración & dosificación , Sobrecarga de Hierro/tratamiento farmacológico , Sideróforos/administración & dosificación , Triazoles/administración & dosificación , Adolescente , Adulto , Benzoatos/efectos adversos , Niño , Deferasirox , Deferoxamina/efectos adversos , Femenino , Corazón/efectos de los fármacos , Humanos , Quelantes del Hierro/efectos adversos , Sobrecarga de Hierro/etiología , Hígado/química , Hígado/efectos de los fármacos , Masculino , Miocardio/química , Sideróforos/efectos adversos , Reacción a la Transfusión , Triazoles/efectos adversos , Adulto Joven
16.
Eur J Haematol ; 92(6): 521-6, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24460655

RESUMEN

OBJECTIVE: Patients with non-transfusion-dependent thalassemia (NTDT) often develop iron overload and related complications, and may require iron chelation. However, the risk of over-chelation emerges as patients reach low, near-normal body iron levels and dose adjustments may be needed. In the THALASSA study, the threshold for chelation interruption was LIC <3 mg Fe/g dw (LIC<3); 24 patients receiving deferasirox for up to 2 yr reached this target. A post hoc analysis was performed to characterize the safety profile of deferasirox as these patients approached LIC<3. METHODS: THALASSA was a randomized, double-blind, placebo-controlled study of two deferasirox regimens (5 and 10 mg/kg/d) versus placebo in patients with NTDT. Patients randomized to deferasirox or placebo in the core could enter a 1-yr extension, with all patients receiving deferasirox (extension starting doses based on LIC at end-of-core and prior chelation response). The deferasirox safety profile was assessed between baseline and 6 months before reaching LIC<3 (Period 1), and the 6 months immediately before achieving LIC<3 (Period 2). RESULTS: Mean ± SD deferasirox treatment duration up to reaching LIC<3 was 476 ± 207 d, and deferasirox dose was 9.7 ± 3.0 mg/kg/d. The exposure-adjusted AE incidence regardless of causality was similar in periods 1 (1.026) and 2 (1.012). There were no clinically relevant differences in renal and hepatic laboratory parameters measured close to the time of LIC<3 compared with measurements near the previous LIC assessment. CONCLUSIONS: The deferasirox safety profile remained consistent as patients approached the chelation interruption target, indicating that, with appropriate monitoring and dose adjustments in relation to iron load, low iron burdens may be reached with deferasirox with minimal risk of over-chelation.


Asunto(s)
Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/metabolismo , Hierro/metabolismo , Hígado/metabolismo , Talasemia/complicaciones , Benzoatos/efectos adversos , Benzoatos/uso terapéutico , Deferasirox , Humanos , Quelantes del Hierro/efectos adversos , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Hígado/patología , Reacción a la Transfusión , Resultado del Tratamiento , Triazoles/efectos adversos , Triazoles/uso terapéutico
17.
Expert Rev Hematol ; 6(5): 495-509, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24083402

RESUMEN

Non-transfusion-dependent thalassemia (NTDT) defines a group of patients who do not require regular transfusions for survival, but are at significant risk of iron accumulation from underlying disease-related mechanisms distinct from transfusional iron overload. Management of iron overload in NTDT has received little attention compared with that of ß-thalassemia major, despite evidence of significant iron-induced complications with advancing age. The efficacy and safety of the iron chelator deferasirox in NTDT has been evaluated in two pilot studies and the first prospective, randomized, placebo-controlled study (THALASSA) of any chelator in NTDT. Treatment with deferasirox for up to 2 years yielded a sustained reduction in iron burden, with a clinically manageable safety profile. Following these trial data, deferasirox is the first iron chelator approved for use in NTDT patients, and with NTDT guidelines now available, physicians are better equipped to achieve effective monitoring and management of iron burden in NTDT.


Asunto(s)
Benzoatos/uso terapéutico , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Talasemia/complicaciones , Triazoles/uso terapéutico , Benzoatos/farmacología , Deferasirox , Humanos , Quelantes del Hierro/farmacología , Talasemia/terapia , Reacción a la Transfusión , Triazoles/farmacología
18.
Ann Hematol ; 92(11): 1485-93, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23775581

RESUMEN

Patients with non-transfusion-dependent thalassemia (NTDT) often develop iron overload that requires chelation to levels below the threshold associated with complications. This can take several years in patients with high iron burden, highlighting the value of long-term chelation data. Here, we report the 1-year extension of the THALASSA trial assessing deferasirox in NTDT; patients continued with deferasirox or crossed from placebo to deferasirox. Of 133 patients entering extension, 130 completed. Liver iron concentration (LIC) continued to decrease with deferasirox over 2 years; mean change was -7.14 mg Fe/g dry weight (dw) (mean dose 9.8 ± 3.6 mg/kg/day). In patients originally randomized to placebo, whose LIC had increased by the end of the core study, LIC decreased in the extension with deferasirox with a mean change of -6.66 mg Fe/g dw (baseline to month 24; mean dose in extension 13.7 ± 4.6 mg/kg/day). Of 166 patients enrolled, 64 (38.6 %) and 24 (14.5 %) patients achieved LIC <5 and <3 mg Fe/g dw by the end of the study, respectively. Mean LIC reduction was greatest in patients with the highest pretreatment LIC. Deferasirox progressively decreases iron overload over 2 years in NTDT patients with both low and high LIC. Safety profile of deferasirox over 2 years was consistent with that in the core study.


Asunto(s)
Benzoatos/uso terapéutico , Transfusión Sanguínea , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Talasemia/tratamiento farmacológico , Triazoles/uso terapéutico , Estudios Cruzados , Deferasirox , Método Doble Ciego , Humanos , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/epidemiología , Estudios Prospectivos , Talasemia/sangre , Talasemia/epidemiología , Factores de Tiempo , Resultado del Tratamiento
19.
Am J Hematol ; 88(6): 503-6, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23553596

RESUMEN

The 1-year THALASSA study enrolled 166 patients with various non-transfusion-dependent thalassemia (NTDT) syndromes, degrees of iron burden and patient characteristics, and demonstrated the overall efficacy and safety of deferasirox in reducing liver iron concentration (LIC) in these patients. Here, reduction in LIC with deferasirox 5 and 10 mg/kg/day starting dose groups is shown to be consistent across the following patient subgroups-baseline LIC/serum ferritin, age, gender, race, splenectomy (yes/no), and underlying NTDT syndrome (ß-thalassemia intermedia, HbE/ß-thalassemia or α-thalassemia). These analyses also evaluated deferasirox dosing strategies for patients with NTDT. Greater reductions in LIC were achieved in patients dose-escalated at Week 24 from deferasirox 10 mg/kg/day starting dose to 20 mg/kg/day. Patients who received an average actual dose of deferasirox >12.5-≤17.5 mg/kg/day achieved a greater LIC decrease compared with the ≥7.5-≤12.5 mg/kg/day and >0-<7.5 mg/kg/day subgroups, demonstrating a dose-response efficacy. LIC reduction across patient subgroups was generally consistent with the primary efficacy analysis with a similar safety profile.


Asunto(s)
Benzoatos/administración & dosificación , Quelantes del Hierro/administración & dosificación , Hígado/efectos de los fármacos , Hígado/metabolismo , Talasemia/tratamiento farmacológico , Talasemia/metabolismo , Triazoles/administración & dosificación , Adulto , Benzoatos/efectos adversos , Deferasirox , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Ferritinas/sangre , Humanos , Quelantes del Hierro/efectos adversos , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/metabolismo , Masculino , Talasemia/sangre , Triazoles/efectos adversos , Adulto Joven
20.
Am J Hematol ; 88(5): 409-15, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23475638

RESUMEN

Despite receiving no or only occasional blood transfusions, patients with non-transfusion-dependent thalassemia (NTDT) have increased intestinal iron absorption and can accumulate iron to levels comparable with transfusion-dependent patients. This iron accumulation occurs more slowly in NTDT patients compared to transfusion-dependent thalassemia patients, and complications do not arise until later in life. It remains crucial for these patients' health to monitor and appropriately treat their iron burden. Based on recent data, including a randomized clinical trial on iron chelation in NTDT, a simple iron chelation treatment algorithm is presented to assist physicians with monitoring iron burden and initiating chelation therapy in this group of patients.


Asunto(s)
Sobrecarga de Hierro/terapia , Talasemia/fisiopatología , Terapia por Quelación/efectos adversos , Humanos , Absorción Intestinal , Quelantes del Hierro/efectos adversos , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/fisiopatología , Hierro de la Dieta/efectos adversos , Hierro de la Dieta/metabolismo , Talasemia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...