Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 280
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Pediatr Hematol Oncol ; 44(1): e26-e34, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34986131

RESUMEN

BACKGROUND: Studies on the increased body iron load in patients with thalassemia major have thoroughly demonstrated the problems caused by iron overload. In patients who undergo hematopoietic stem cell transplantation (HSCT) as curative therapy, iron overload continues long after transplantation. There are few pediatric studies on chelation therapy in the posttransplant period. In this study, we present the outcomes of our patients who received posttransplant oral chelation therapy. PATIENTS AND METHODS: This retrospective observational study evaluated the outcomes of pediatric patients with thalassemia major who used oral chelation therapy after allogeneic HSCT at the Akdeniz University Pediatric Bone Marrow Unit between January 2008 and October 2019. RESULTS: Deferasirox therapy was initiated in 58 pediatric patients who underwent HSCT for thalassemia. Pretreatment mean serum ferritin was 2166±1038 ng/mL. Treatment was initiated at a mean of 12±6.7 months after transplantation and continued for a mean of 15.7±11.5 months. At treatment discontinuation, the mean serum ferritin was 693±405 ng/mL and the mean reduction was -1472.75±1121.09 ng/mL (P<0.001 vs. posttreatment). Serum ferritin was below 500 ng/mL in 52% of the patients at treatment discontinuation. Manageable side effects such as nausea, vomiting, liver enzyme elevation, and proteinuria were observed in 17% of the patients, while one patient developed ototoxicity. CONCLUSIONS: Deferasirox therapy effectively reduces iron overload in the posttransplant period. Studies evaluating the effects of early treatment on the graft may help to establish guidelines for posttransplant chelation therapy. Clear guidelines are needed regarding when to initiate and discontinue treatment.


Asunto(s)
Deferasirox/administración & dosificación , Trasplante de Células Madre Hematopoyéticas , Quelantes del Hierro/administración & dosificación , Sobrecarga de Hierro/terapia , Talasemia/terapia , Adolescente , Aloinjertos , Niño , Preescolar , Deferasirox/efectos adversos , Femenino , Ferritinas/sangre , Humanos , Quelantes del Hierro/efectos adversos , Sobrecarga de Hierro/sangre , Masculino , Estudios Retrospectivos , Talasemia/sangre
2.
Biomed Pharmacother ; 145: 112381, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34736078

RESUMEN

The oxidative status of twenty-three ß-thalassemia/hemoglobin E patients was evaluated after administration of 75 mg/kg deferiprone (GPO-L-ONE®) divided into 3 doses daily for 12 months. Serum ferritin was significantly decreased; the median value at the initial and final assessments was 2842 and 1719 ng/mL, respectively. Progressive improvement with significant changes in antioxidant enzyme activity, including plasma paraoxonase (PON) and platelet-activating factor acetylhydrolase (PAF-AH), and in antioxidant enzymes in red blood cells (glutathione peroxidase (GPx), catalase and superoxide dismutase (SOD)) were observed at 3-6 months of treatment. The levels of total GSH in red blood cells were significantly increased at the end of the study. Improved red blood cell membrane integrity was also demonstrated using the EPR spin labeling technique. Membrane fluidity at the surface and hydrophobic regions of the red blood cell membrane was significantly changed after 12 months of treatment. In addition, a significant increase in hemoglobin content was observed (6.6 ± 0.7 and 7.5 ± 1.3 g/dL at the initial assessment and at 6 months, respectively). Correlations were observed between hemoglobin content, membrane fluidity and antioxidant enzymes in red blood cells. The antioxidant activity of deferiprone may partly be explained by progressive reduction of redox active iron that catalyzes free radical reactions, as demonstrated by the EPR spin trapping technique. In conclusion, iron chelation therapy with deferiprone notably improved the oxidative status in thalassemia, consequently reducing the risk of oxidative-related complications. Furthermore, the improvement in red blood cell quality may improve the anemia situation in patients.


Asunto(s)
Deferiprona/farmacología , Quelantes del Hierro/farmacología , Hierro/metabolismo , Talasemia beta/tratamiento farmacológico , Adolescente , Adulto , Antioxidantes/metabolismo , Deferiprona/administración & dosificación , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Femenino , Ferritinas/sangre , Glutatión Peroxidasa/metabolismo , Hemoglobina E/metabolismo , Humanos , Quelantes del Hierro/administración & dosificación , Masculino , Persona de Mediana Edad , Oxidación-Reducción , Superóxido Dismutasa/metabolismo , Adulto Joven
3.
Food Funct ; 12(21): 10887-10902, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34643632

RESUMEN

Iron deficiency is a global nutritional problem that adversely affects the functional regulation of the immune system. In the process of treatment through iron supplementation, the problem of excessive iron intake often occurs, which increases the level of inflammation in the body. Excessive iron can also lead to an increase in intestinal iron-requiring pathogenic bacteria and an imbalance of intestinal flora. In this study, we aim to explore the effect of Ejiao peptide-iron (EPI) chelates on the intestinal flora and inflammation of ICR mice having iron-deficiency anemia (IDA). The mice were given low, medium, and high doses of EPI and FeSO4 (1.0, 2.0 and 3.0 mg Fe per kg weight, respectively) daily for 4 weeks by intragastric administration. IDA mice showed increased inflammation levels and decreased sIgA secretion, which were restored after intervention with EPI at different doses. Intestinal mucosal ulcers, inflammatory cell infiltration, and oxidative stress in the colon tissue were reduced, and intestinal permeability was improved. Furthermore, 16S rDNA gene sequencing revealed that EPI increased microbial diversity and richness, changing the community structure, therefore, alleviating microbiota dysbiosis caused by IDA (e.g. the proportion of Firmicutes/Bacteroides). Different from the traditional iron supplement FeSO4, when the pathogenic bacteria (e.g. Helicobacter and Erysipelatoclostridium) increase and the beneficial bacteria (e.g. Bifidobacterium and Blautia) decrease at high doses, EPI shows higher safety at a high dose, thereby maintaining a healthier intestinal homeostasis.


Asunto(s)
Anemia Ferropénica/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Gelatina/uso terapéutico , Inflamación/tratamiento farmacológico , Enfermedades Intestinales/tratamiento farmacológico , Quelantes del Hierro/uso terapéutico , Animales , Bacterias/genética , Biomarcadores , Regulación de la Expresión Génica/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento , Quelantes del Hierro/administración & dosificación , Masculino , Ratones , Ratones Endogámicos ICR , Estrés Oxidativo/efectos de los fármacos , ARN/genética , ARN/metabolismo , ARN Bacteriano , ARN Ribosómico 16S
4.
J Ayub Med Coll Abbottabad ; 33(2): 207-212, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34137530

RESUMEN

BACKGROUND: Thalassemia major is the severe form of ß thalassemia characterized by severe anaemia, hepatosplenomegaly and facioskeletal changes due to increased haemolysis of defective red blood cells. In iron overload states, high levels of iron exceed the iron-carrying capacity of transferrin within the plasma, leading to the formation of nontransferrin-bound iron form. These nontransferrin-bound iron forms can be taken up into cells, including liver, heart, and endocrine cells leading to organ damage. To prevent complications associated with hemosiderosis, iron chelation therapy remains one of the main objectives of clinical management of the patients affected by Thalassemia Major. METHODS: Thirty-seven patients were enrolled using non randomized convenience sampling technique after the written consent from patients. Patients age 2-30 years were enrolled in this study. Serum Ferritin, ALT, Serum Creatinine were checked at the start of the study, 3 months, 6months and then at the end of the study, i.e., at 9 months of the commencement of the study. They were also assessed for other side effects pertaining to oral tolerability of the drug like vomiting, nausea, GI upset, diarrhoea, urinary complaints or any other subjective complaint. RESULTS: Of the 37 patients, 20 were male (54.1%) and 17 were female (45.9%). Mean age of the patients was 10.2 years (Min. 3 years, Max 21 years). The average serum Ferritin at baseline was noted as 3440 which increased after a period of 3 months, 6 months and 9 months with average of 3359, 3677 and 4394 respectively. After the period of 9 months largest 95% confidence interval of serum Ferritin levels was observed in the range of 3420.17 to 5368.63. In our study, 17 patients required alternative chelation (46%). These patients needed IV Deferioxamine because of the rising trend of Serum Ferritin after the study. CONCLUSIONS: From the results of our study, we infer that oral Deferasirox is not an effective iron chelator. If the patients are taking oral deferasirox, their Serum Ferritin should be checked 3 monthlies. The drug is effective only in maintaining Serum Ferritin levels with levels less than 1500ng/ml. Intravenous Deferioxamine still should be preferred over oral iron chelators for effective control of iron overload and its complications.


Asunto(s)
Deferasirox/uso terapéutico , Quelantes del Hierro/uso terapéutico , Talasemia beta/tratamiento farmacológico , Administración Intravenosa , Adolescente , Adulto , Benzoatos/efectos adversos , Niño , Preescolar , Deferasirox/administración & dosificación , Deferasirox/efectos adversos , Recuento de Eritrocitos , Femenino , Hepatomegalia , Humanos , Quelantes del Hierro/administración & dosificación , Quelantes del Hierro/efectos adversos , Masculino , Triazoles/efectos adversos , Adulto Joven
5.
Sci Rep ; 11(1): 12581, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131221

RESUMEN

Deferasirox (DFX) is the newest among three different chelators available to treat iron overload in iron-loading anaemias, firstly released as Dispersible Tablets (DT) and more recently replaced by Film-Coated Tablets (FCT). In this retrospective observational study, pharmacokinetics, pharmacodynamics, and safety features of DFX treatment were analyzed in 74 patients that took both formulations subsequently under clinical practice conditions. Bioavailability of DFX FCT compared to DT resulted higher than expected [Cmax: 99.5 (FCT) and 69.7 (DT) µMol/L; AUC: 1278 (FCT) and 846 (DT), P < 0.0001]. DFX FCT was also superior in scalability among doses. After one year of treatment for each formulation, no differences were observed between the treatments in the overall iron overload levels; however, DFX FCT but not DT showed a significant dose-response correlation [Spearman r (dose-serum ferritin variation): - 0.54, P < 0.0001]. Despite being administered at different dosages, the long-term safety profile was not different between formulations: a significant increase in renal impairment risk was observed for both treatments and it was reversible under strict monitoring (P < 0.002). Altogether, these data constitute a comprehensive comparison of DFX formulations in thalassaemia and other iron-loading anaemias, confirming the effectiveness and safety characteristics of DFX and its applicability for treatment tailoring.


Asunto(s)
Anemia/tratamiento farmacológico , Deferasirox/administración & dosificación , Sobrecarga de Hierro/tratamiento farmacológico , Talasemia/tratamiento farmacológico , Adulto , Anemia/sangre , Anemia/epidemiología , Anemia/patología , Terapia por Quelación/tendencias , Deferasirox/farmacocinética , Femenino , Ferritinas/sangre , Humanos , Hierro/sangre , Hierro/metabolismo , Quelantes del Hierro/administración & dosificación , Quelantes del Hierro/farmacocinética , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/epidemiología , Sobrecarga de Hierro/patología , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Talasemia/sangre , Talasemia/epidemiología , Talasemia/patología
6.
Expert Opin Drug Saf ; 20(8): 937-947, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33877003

RESUMEN

Introduction: Beta-thalassemia is an autosomal recessive hereditary anemia characterized by reduced or absent ß-globin chain synthesis, affecting about 60,000 people peryear. Management for ß-thalassemia major includes regular blood transfusions followed by iron chelating therapy and drug targeting ineffective erythropoiesis.Areas covered: The safety of licensed drugs for the management of ß-thalassemia is reviewed, using evidence from clinical trials and observational research. Such drugs include the iron chelators and the erythrocyte maturation agent luspatercept. The safety of emerging treatment, such as hydroxyurea and thalidomide is also reviewed.Expert opinion: Beta-thalassemia is arare disease, and is not surprising that there are limited studies investigating the safety of drugs used in this disease. Indeed, although observational studies are the main source of drug safety information in areal-world setting, only eleven studies were identified for iron-chelators and none of these estimated the risk of agiven safety outcome. Future work should aim to better leverage existing sources of real-world datato investigate drug safety in thalassemia.


Asunto(s)
Receptores de Activinas Tipo II/efectos adversos , Fragmentos Fc de Inmunoglobulinas/efectos adversos , Quelantes del Hierro/efectos adversos , Proteínas Recombinantes de Fusión/efectos adversos , Talasemia beta/tratamiento farmacológico , Receptores de Activinas Tipo II/administración & dosificación , Hematínicos/administración & dosificación , Hematínicos/efectos adversos , Humanos , Hidroxiurea/administración & dosificación , Hidroxiurea/efectos adversos , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Quelantes del Hierro/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Talidomida/administración & dosificación , Talidomida/efectos adversos , Talasemia beta/fisiopatología
7.
Am J Clin Nutr ; 114(2): 780-793, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-33871598

RESUMEN

BACKGROUND: The iron-chelating activities of polyphenols raise concern whether there is a risk of iron deficiency or anemia induced by polyphenol supplementation. Results from clinical trials regarding the effects of polyphenol supplementation on iron status and erythropoiesis are inconclusive. OBJECTIVE: We performed a systematic review and meta-analysis of randomized controlled trials to determine the effects of polyphenol supplementation on iron status and erythropoiesis. METHODS: Published articles were searched between May 1988 and 7 December, 2020. Finally, we identified 34 randomized controlled trials. Random-effects meta-analyses were performed to obtain the weighted mean difference of serum iron (SI), transferrin saturation (TS), ferritin, and hemoglobin concentration. Funnel plots and Egger's test were used to determine the risk of bias. The robustness of the effect sizes was examined by sensitivity analysis. RESULTS: Polyphenol supplementation had an inhibitory effect on the SI concentration (-13.72 µg/dL; 95% CI: -20.74, -6.71) and TS (-3.10%; 95% CI: -4.93, -1.27), with no effect on ferritin (-9.34 ng/mL; 95% CI: -28.55, 9.87). Polyphenols increased the hemoglobin concentration (8.53 g/L; 95% CI: 3.33, 13.73). In healthy participants, polyphenol reduced the TS (-3.83%; 95% CI: -7.47, -0.19) and increased the hemoglobin concentration (12.87 g/L; 95% CI: 1.61, 24.14). Similarly, polyphenol reduced the SI concentration (-8.60 µg/dL; 95% CI: -16.10, -1.10) and increased the hemoglobin concentration (8.50 g/L; 95% CI: 0.86, 16.15) in patients with metabolic diseases. In patients with ß-thalassemia, polyphenol decreased the SI concentration (-23.19 µg/dL; 95% CI: -35.84, -10.55), TS (-3.23%; 95% CI: -5.54, -0.91), and ferritin concentration (-223.62 ng/mL; 95% CI: -359.32, -87.91), but had no effect on the hemoglobin concentration. CONCLUSION: Healthy individuals and patients with metabolic diseases may benefit from the positive impact of polyphenols on erythropoiesis. Patients with ß-thalassemia may benefit from the effect of polyphenols on reducing SI. This trial was registered at PROSPERO (International prospective register of systematic reviews) as CRD42020161983.


Asunto(s)
Anemia Ferropénica/inducido químicamente , Eritropoyesis/efectos de los fármacos , Quelantes del Hierro/administración & dosificación , Hierro/metabolismo , Polifenoles/administración & dosificación , Suplementos Dietéticos , Humanos , Quelantes del Hierro/efectos adversos , Polifenoles/efectos adversos
8.
PLoS One ; 16(4): e0249527, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33822801

RESUMEN

OBJECTIVE: A meta-analysis was conducted to assess the effects of dietary chromium picolinate (CrPic) supplementation on broiler growth performance and to determine whether such effects are regulated by broiler strains, sex, environmental stress, or contextual factors including study area and years. METHODS: Eligible studies were identified by searching the Web of Science, Springer, Elsevier, ScienceDirect, Taylor & Francis Online databases. Weighted average differences with corresponding 95% confidence intervals were computed with a random-effects model. We performed subgroup analysis stratified by study area, published years, broiler strains and sex, and environmental stress. Publication bias was assessed with Egger's test method. A total of 15 studies eligible for inclusion. RESULTS: The results indicated that CrPic supplementation significantly improved broiler growth performance and subgroup analysis confirmed this conclusion. We also found that Ross 308 or male broilers might be more sensitive to CrPic supplementation and showed better growth performance. A model was used to obtain the amount of chromium addition under the optimal growth performance, which suggested that the maximum value of average daily gain (ADG) was reached when chromium addition was 1810 µg/kg. The results of the sensitivity analysis showed low sensitivity and high stability of the meta-analysis. CONCLUSIONS: CrPic supplementation had a positive effect on the growth performance of broilers, and this meta-analysis provides a more accurate value of chromium addition, which may be beneficial for the practice of the broiler industry.


Asunto(s)
Alimentación Animal/análisis , Pollos/crecimiento & desarrollo , Suplementos Dietéticos , Quelantes del Hierro/administración & dosificación , Ácidos Picolínicos/administración & dosificación , Animales
9.
Int J Mol Sci ; 22(2)2021 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-33467196

RESUMEN

In ß-thalassemia, ineffective erythropoiesis leads to anemia and systemic iron overload. The management of iron overload by chelation therapy is a standard of care. However, iron chelation does not improve the ineffective erythropoiesis. We recently showed that the oral ferroportin inhibitor VIT-2763 ameliorates anemia and erythropoiesis in the Hbbth3/+ mouse model of ß-thalassemia. In this study, we investigated whether concurrent use of the iron chelator deferasirox (DFX) and the ferroportin inhibitor VIT-2763 causes any pharmacodynamic interactions in the Hbbth3/+ mouse model of ß-thalassemia. Mice were treated with VIT-2763 or DFX alone or with the combination of both drugs once daily for three weeks. VIT-2763 alone or in combination with DFX improved anemia and erythropoiesis. VIT-2763 alone decreased serum iron and transferrin saturation (TSAT) but was not able to reduce the liver iron concentration. While DFX alone had no effect on TSAT and erythropoiesis, it significantly reduced the liver iron concentration alone and in the presence of VIT-2763. Our results clearly show that VIT-2763 does not interfere with the iron chelation efficacy of DFX. Furthermore, VIT-2763 retains its beneficial effects on improving ineffective erythropoiesis when combined with DFX in the Hbbth3/+ mouse model. In conclusion, co-administration of the oral ferroportin inhibitor VIT-2763 and the iron chelator DFX is feasible and might offer an opportunity to improve both ineffective erythropoiesis and iron overload in ß-thalassemia.


Asunto(s)
Bencimidazoles/farmacología , Eritropoyesis/efectos de los fármacos , Oxazoles/farmacología , Piridinas/farmacología , Talasemia beta/tratamiento farmacológico , Administración Oral , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/efectos adversos , Bencimidazoles/uso terapéutico , Proteínas de Transporte de Catión/antagonistas & inhibidores , Células Cultivadas , Deferasirox/administración & dosificación , Deferasirox/farmacología , Deferasirox/uso terapéutico , Combinación de Medicamentos , Interacciones Farmacológicas , Femenino , Hierro/sangre , Quelantes del Hierro/administración & dosificación , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Oxazoles/administración & dosificación , Oxazoles/efectos adversos , Oxazoles/uso terapéutico , Piridinas/administración & dosificación , Piridinas/efectos adversos , Piridinas/uso terapéutico , Transferrina/metabolismo
10.
Qual Life Res ; 30(2): 467-477, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32920766

RESUMEN

PURPOSE: To evaluate health-related quality of life (HRQoL) and satisfaction with iron chelation therapy (ICT) of patients with transfusion-dependent ß-thalassemia (TDT) managed under routine care conditions. PATIENTS AND METHODS: This was an observational, multicenter, cross-sectional study conducted in three hospital-based Thalassemia Units of Western Greece. Patients confidentially completed the 36-item short-form (SF-36) and the "satisfaction with ICT" (SICT) instruments to assess HRQoL and ICT satisfaction respectively. RESULTS: One hundred and thirty-one adult TDT patients [74 female, median (IQR) age: 41 (36-47) years] were enrolled. Eighty patients (61.1%) were receiving parenteral ICT, with or without oral chelators (Group I), whereas 51 (38.9%) were only receiving oral ICT (Group II). The median SF-36 physical component summary and mental component summary scores were 76.3 and 75.7 among Group I, and 76.9 and 74.5 among Group II patients, not differing between the two groups. In their majority, Group I (84.6%) and Group II (92.9%) patients reported preferring oral ICT. Moreover, Group I patients reported greater perceived ICT effectiveness (median SICT score: 4.3 versus 4.2; p = 0.039), whereas patients receiving deferasirox-containing ICT reported higher treatment acceptance (median SICT score: 4.0 versus 3.6, p = 0.038) and greater satisfaction with the burden of their ICT (median SICT score: 4.4 versus 3.9, p = 0.033). CONCLUSION: TDT patients prefer to receive oral ICT and are more satisfied of the burden of deferasirox-containing ICT, even though those receiving parenteral ICT are more satisfied by the effectiveness of their treatment. No differences in HRQoL were not noted between patients receiving parenteral versus oral ICT.


Asunto(s)
Quelantes del Hierro/uso terapéutico , Talasemia beta/tratamiento farmacológico , Adulto , Estudios Transversales , Femenino , Grecia , Humanos , Quelantes del Hierro/administración & dosificación , Masculino , Satisfacción del Paciente , Calidad de Vida/psicología
11.
Int J Mol Sci ; 21(22)2020 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-33233561

RESUMEN

Thalassemia syndromes are characterized by the inability to produce normal hemoglobin. Ineffective erythropoiesis and red cell transfusions are sources of excess iron that the human organism is unable to remove. Iron that is not saturated by transferrin is a toxic agent that, in transfusion-dependent patients, leads to death from iron-induced cardiomyopathy in the second decade of life. The availability of effective iron chelators, advances in the understanding of the mechanism of iron toxicity and overloading, and the availability of noninvasive methods to monitor iron loading and unloading in the liver, heart, and pancreas have all significantly increased the survival of patients with thalassemia. Prolonged exposure to iron toxicity is involved in the development of endocrinopathy, osteoporosis, cirrhosis, renal failure, and malignant transformation. Now that survival has been dramatically improved, the challenge of iron chelation therapy is to prevent complications. The time has come to consider that the primary goal of chelation therapy is to avoid 24-h exposure to toxic iron and maintain body iron levels within the normal range, avoiding possible chelation-related damage. It is very important to minimize irreversible organ damage to prevent malignant transformation before complications set in and make patients ineligible for current and future curative therapies. In this clinical case-based review, we highlight particular aspects of the management of iron overload in patients with beta-thalassemia syndromes, focusing on our own experience in treating such patients. We review the pathophysiology of iron overload and the different ways to assess, quantify, and monitor it. We also discuss chelation strategies that can be used with currently available chelators, balancing the need to keep non-transferrin-bound iron levels to a minimum (zero) 24 h a day, 7 days a week and the risk of over-chelation.


Asunto(s)
Deferoxamina/administración & dosificación , Quelantes del Hierro/administración & dosificación , Sobrecarga de Hierro/tratamiento farmacológico , Hierro/metabolismo , Reacción a la Transfusión/complicaciones , Talasemia beta/terapia , Adulto , Transfusión Sanguínea , Cardiomiopatías/sangre , Cardiomiopatías/etiología , Cardiomiopatías/fisiopatología , Cardiomiopatías/prevención & control , Terapia por Quelación/efectos adversos , Terapia por Quelación/métodos , Deferoxamina/efectos adversos , Monitoreo de Drogas/instrumentación , Monitoreo de Drogas/métodos , Femenino , Corazón/efectos de los fármacos , Corazón/fisiopatología , Humanos , Hierro/toxicidad , Quelantes del Hierro/efectos adversos , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/fisiopatología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Persona de Mediana Edad , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Páncreas/patología , Transferrina/metabolismo , Reacción a la Transfusión/sangre , Reacción a la Transfusión/fisiopatología , Talasemia beta/metabolismo , Talasemia beta/patología
13.
Ann Hematol ; 99(10): 2289-2294, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32737633

RESUMEN

Iron overload-induced cardiomyopathy is the leading cause of death in patients with transfusion-dependent thalassemia (TDT). The mortality is extremely high in these patients with severe cardiac complications, and how to rescue them remains a challenge. It is reasonable to use combined chelation with deferiprone (L1) and deferoxamine (DFO) because of their shuttle and synergistic effects on iron chelation. Here, seven consecutive patients with TDT who had severe cardiac complications between 2002 and 2019 and received combined chelation therapy with oral high-dose L1 (100 mg/kg/day) and continuous 24-h DFO infusion (50 mg/kg/day) in our hospital were reported. Survival for eight consecutive patients receiving DFO monotherapy for their severe cardiac complications between 1984 and 2001 was compared. We found that combined chelation therapy with high-dose L1 and DFO was efficient to improve survival and cardiac function in patients with TDT presenting severe cardiac complications. Reversal of arrhythmia to sinus rhythm was noted in all patients. Their 1-month follow-up left ventricular ejection fraction increased significantly (P < 0.001). There were no deaths, and all patients were discharged from hospital with good quality of life. In contrast, all the eight patients receiving DFO monotherapy died (P < 0.001). Accordingly, combined chelation therapy with high-dose L1 and DFO should be considered in patients with TDT presenting cardiac complications.


Asunto(s)
Arritmias Cardíacas/tratamiento farmacológico , Terapia por Quelación/métodos , Deferiprona/uso terapéutico , Deferoxamina/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Talasemia/terapia , Arritmias Cardíacas/etiología , Arritmias Cardíacas/fisiopatología , Transfusión Sanguínea , Deferiprona/administración & dosificación , Deferoxamina/administración & dosificación , Evaluación de Medicamentos , Quimioterapia Combinada , Femenino , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/fisiopatología , Humanos , Quelantes del Hierro/administración & dosificación , Sobrecarga de Hierro/etiología , Masculino , Calidad de Vida , Estudios Retrospectivos , Talasemia/complicaciones , Reacción a la Transfusión , Resultado del Tratamiento , Función Ventricular Izquierda
14.
Lancet Haematol ; 7(6): e469-e478, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32470438

RESUMEN

BACKGROUND: Transfusion-dependent haemoglobinopathies require lifelong iron chelation therapy with one of the three iron chelators (deferiprone, deferasirox, or deferoxamine). Deferasirox and deferiprone are the only two oral chelators used in adult patients with transfusion-dependent haemoglobinopathies. To our knowledge, there are no randomised clinical trials comparing deferiprone, a less expensive iron chelator, with deferasirox in paediatric patients. We aimed to show the non-inferiority of deferiprone versus deferasirox. METHODS: DEEP-2 was a phase 3, multicentre, randomised trial in paediatric patients (aged 1 month to 18 years) with transfusion-dependent haemoglobinopathies. The study was done in 21 research hospitals and universities in Italy, Egypt, Greece, Albania, Cyprus, Tunisia, and the UK. Participants were receiving at least 150 mL/kg per year of red blood cells for the past 2 years at the time of enrolment, and were receiving deferoxamine (<100 mg/kg per day) or deferasirox (<40 mg/kg per day; deferasirox is not registered for use in children aged <2 years so only deferoxamine was being used in these patients). Any previous chelation treatment was permitted with a 7-day washout period. Patients were randomly assigned 1:1 to receive orally administered daily deferiprone (75-100 mg/kg per day) or daily deferasirox (20-40 mg/kg per day) administered as dispersible tablets, both with dose adjustment for 12 months, stratified by age (<10 years and ≥10 years) and balanced by country. The primary efficacy endpoint was based on predefined success criteria for changes in serum ferritin concentration (all patients) and cardiac MRI T2-star (T2*; patients aged >10 years) to show non-inferiority of deferiprone versus deferasirox in the per-protocol population, defined as all randomly assigned patients who received the study drugs and had available data for both variables at baseline and after 1 year of treatment, without major protocol violations. Non-inferiority was based on the two-sided 95% CI of the difference in the proportion of patients with treatment success between the two groups and was shown if the lower limit of the two-sided 95% CI was greater than -12·5%. Safety was assessed in all patients who received at least one dose of study drug. This study is registered with EudraCT, 2012-000353-31, and ClinicalTrials.gov, NCT01825512. FINDINGS: 435 patients were enrolled between March 17, 2014, and June 16, 2016, 393 of whom were randomly assigned to a treatment group (194 to the deferiprone group; 199 to the deferasirox group). 352 (90%) of 390 patients had ß-thalassaemia major, 27 (7%) had sickle cell disease, five (1%) had thalassodrepanocytosis, and six (2%) had other haemoglobinopathies. Median follow-up was 379 days (IQR 294-392) for deferiprone and 381 days (350-392) for deferasirox. Non-inferiority of deferiprone versus deferasirox was established (treatment success in 69 [55·2%] of 125 patients assigned deferiprone with primary composite efficacy endpoint data available at baseline and 1 year vs 80 [54·8%] of 146 assigned deferasirox, difference 0·4%; 95% CI -11·9 to 12·6). No significant difference between the groups was shown in the occurrence of serious and drug-related adverse events. Three (2%) cases of reversible agranulocytosis occurred in the 193 patients in the safety analysis in the deferiprone group and two (1%) cases of reversible renal and urinary disorders (one case of each) occurred in the 197 patients in the deferasirox group. Compliance was similar between treatment groups: 183 (95%) of 193 patients in the deferiprone group versus 192 (97%) of 197 patients in the deferisirox group. INTERPRETATION: In paediatric patients with transfusion-dependent haemoglobinopathies, deferiprone was effective and safe in inducing control of iron overload during 12 months of treatment. Considering the need for availability of more chelation treatments in paediatric populations, deferiprone offers a valuable treatment option for this age group. FUNDING: EU Seventh Framework Programme.


Asunto(s)
Deferasirox/uso terapéutico , Deferiprona/uso terapéutico , Transfusión de Eritrocitos/métodos , Hemoglobinopatías/tratamiento farmacológico , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Administración Oral , Adolescente , Agranulocitosis/inducido químicamente , Agranulocitosis/epidemiología , Albania/epidemiología , Anemia de Células Falciformes/terapia , Técnicas de Imagen Cardíaca/métodos , Niño , Preescolar , Chipre/epidemiología , Deferasirox/administración & dosificación , Deferasirox/economía , Deferiprona/administración & dosificación , Deferiprona/economía , Egipto/epidemiología , Transfusión de Eritrocitos/estadística & datos numéricos , Femenino , Ferritinas/sangre , Ferritinas/efectos de los fármacos , Grecia/epidemiología , Hemoglobinopatías/terapia , Humanos , Lactante , Quelantes del Hierro/administración & dosificación , Quelantes del Hierro/economía , Sobrecarga de Hierro/sangre , Italia/epidemiología , Imagen por Resonancia Magnética , Masculino , Cooperación del Paciente , Resultado del Tratamiento , Túnez/epidemiología , Reino Unido/epidemiología , Enfermedades Urológicas/inducido químicamente , Enfermedades Urológicas/epidemiología , Talasemia beta/terapia
15.
Horm Metab Res ; 52(3): 194-201, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32215890

RESUMEN

Derangements in phosphate and calcium homeostasis are common in patients with beta-thalassemia. Fibroblast growth factor 23 (FGF23) is among the main hormones regulating phosphate levels, while several studies underline an interplay between iron (Fe) and FGF23. Herein, we investigated, for the first time, the serum intact molecule (iFGF23) and the carboxyl-terminal fragment (C-FGF23) and Klotho levels simultaneously in patients with beta-thalassemia major receiving iron chelation regimens in comparison to healthy control subjects. We also correlated them with the body iron burden. The observational case-control study included 81 subjects (40 thalassemic patients and 41 healthy controls). Serum iFGF23, C-FGF23 and Κlotho were measured by ELISA. Parathormone, 25-hydroxycholecalciferol, calcium, and phosphorus were measured in blood and/or urine. The degree of hemosiderosis was evaluated by assessing the serum ferritin levels and performing T2* MRI measurements. Serum C-FGF23 levels were significantly lower in patients compared to control subjects (p=0.04), while iFGF23 and Klotho levels did not differ. Serum C-FGF23 levels were negatively correlated with ferritin (r=-0,421, p=0.018), whereas there were no significant correlations of each of the three factors with the iron chelation therapy. Decreased serum C-FGF23 levels were found in ßTh patients which may be attributed to inhibition of proteolytic cleavage of iFGF23. Further studies in a greater number of patients will shed more light on the disturbances of the iFGF23, Klotho and C-FGF23 in thalassemia and their possible role in bone disease of such patients.


Asunto(s)
Factores de Crecimiento de Fibroblastos/sangre , Glucuronidasa/sangre , Talasemia beta/sangre , Adolescente , Adulto , Femenino , Ferritinas/sangre , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Humanos , Hierro/sangre , Quelantes del Hierro/administración & dosificación , Proteínas Klotho , Masculino , Persona de Mediana Edad , Adulto Joven , Talasemia beta/tratamiento farmacológico
16.
Hematol Oncol Clin North Am ; 34(2): 465-473, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32089223

RESUMEN

Iron overload (IOL) in patients with myelodysplastic syndromes (MDS) is mainly attributable to chronic transfusion therapy. The importance of iron chelation therapy (ICT) in MDS has been a matter of debate. The Telesto study, the only randomized, placebo-controlled trial of ICT with deferasirox in MDS, showed improved event-free survival with ICT in patients with lower-risk MDS. Although Telesto was not powered to detect differences between deferasirox and placebo for single-event categories of the composite primary endpoint for event-free survival, results are consistent with the view that iron-related cardiac dysfunction is ameliorated by ICT in elderly patients with MDS.


Asunto(s)
Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Síndromes Mielodisplásicos/complicaciones , Biomarcadores , Transfusión Sanguínea , Terapia por Quelación , Humanos , Quelantes del Hierro/administración & dosificación , Quelantes del Hierro/efectos adversos , Sobrecarga de Hierro/diagnóstico , Síndromes Mielodisplásicos/mortalidad , Síndromes Mielodisplásicos/terapia , Pronóstico , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
17.
Expert Opin Drug Metab Toxicol ; 16(3): 179-193, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32067512

RESUMEN

Background: Deferasirox is the first line of treatment in iron overload. In spite of the many studies concerning the efficacy of deferasirox, some patients remain unresponsive to deferasirox.Methods: One hundred and sixty patients were enrolled in stratified-randomized controlled study. Patients were randomly divided into four regimens, group I (n = 40) received 30 mg/kg deferasirox, group II (n = 40) received 20 mg omeprazole and 30 mg/kg deferasirox, group III (n = 40) received 400 mg vitamin E and 30 mg/kg deferasirox and group IV (n = 40) received 420 mg silymarin and 30 mg/kg deferasirox. Blood specimens were collected from each patient for up to 24 h, and then plasma deferasirox concentrations were inspected.Results: Silymarin, Vitamin E, and omeprazole significantly increased the peak plasma concentration of deferasirox (P < 0.001) by 27.9, 14.9 and 2.4 fold, respectively, as compared to deferasirox alone. The bioavailability of deferasirox was improved up to 3.03, 3.57, and 4.98-fold, respectively, following administration of omeprazole, vitamin E, and silymarin compared to deferasirox alone.Conclusion: Silymarin, vitamin E, and omeprazole represent promising adjuvant therapy to improve the chelation efficacy of deferasirox that might also be further applied to enhance the pharmacokinetics of deferasirox to overcome the lack of response.


Asunto(s)
Transfusión Sanguínea/tendencias , Deferasirox/administración & dosificación , Quelantes del Hierro/administración & dosificación , Talasemia beta/diagnóstico , Talasemia beta/terapia , Adolescente , Niño , Terapia Combinada/tendencias , Deferasirox/sangre , Quimioterapia Combinada , Femenino , Humanos , Quelantes del Hierro/metabolismo , Masculino , Omeprazol/administración & dosificación , Omeprazol/sangre , Resultado del Tratamiento , Vitamina E/administración & dosificación , Vitamina E/sangre , Talasemia beta/sangre
18.
Biometals ; 33(2-3): 87-95, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32100150

RESUMEN

Niemann-Pick Disease Type C (NP-C) is a fatal lysosomal storage disorder with progressive neurodegeneration. In addition to the characteristic cholesterol and lipid overload phenotype, we previously found that altered metal homeostasis is also a pathological feature. Increased brain iron in the Npc1-/- mouse model of NP-C may potentially contribute to neurodegeneration, similar to neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. Deferiprone (DFP) is a brain penetrating iron chelator that has demonstrated effectiveness in preventing neurological deterioration in Parkinson's disease clinical trials. Therefore, we hypothesized that DFP treatment, targeting brain iron overload, may have therapeutic benefits for NP-C. Npc1-/- mice were assigned to four experimental groups: (1) pre-symptomatic (P15) + 75 mg/kg DFP; (2) pre-symptomatic (P15) + 150 mg/kg DFP; (3) symptomatic (P49) + 75 mg/kg DFP; (4) symptomatic (P49) + 150 mg/kg DFP. Our study found that in Npc1-/- mice, DFP treatment did not offer any improvement over the expected disease trajectory and median lifespan. Moreover, earlier treatment and higher dose of DFP resulted in adverse effects on body weight and onset of ataxia. The outcome of our study indicated that, despite increased brain iron, Npc1-/- mice were vulnerable to pharmacological iron depletion, especially in early life. Therefore, based on the current model, iron chelation therapy is not a suitable treatment option for NP-C.


Asunto(s)
Deferiprona/administración & dosificación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Quelantes del Hierro/administración & dosificación , Animales , Modelos Animales de Enfermedad , Femenino , Inyecciones Intraperitoneales , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteína Niemann-Pick C1
19.
Indian J Pharmacol ; 52(6): 514-519, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33666193

RESUMEN

Chronic iron overload in beta-thalassemia patients after continuous blood transfusions has caused notable morbidity and mortality in these patients. The once-a-day oral iron chelator, deferasirox has established efficacy and bearable safety in adults and pediatric thalassemia patients. It is now extensively used for the management of transfusional hemosiderosis. However, a number of studies have revealed a few patients continued to be none respondent or intolerant toward the once-a-day regimen of deferasirox even after the administration of maximum dose recommended by the World Health Organization. In the literature, there were three studies showing the boon of twice in a day dosing of deferasirox among transfusional-dependent beta thalassemia patients. Therefore, a nonsystematic review was conducted on above three studies to ascertain the enhanced effectiveness and tolerability of twice per day regimen of deferasirox with the same total dose as that of once daily regimen of deferasirox in unresponsive or intolerant transfusion-dependent beta-thalassemia (TDT) patients. All the above studies concluded that the twice per day regimen of deferasirox was more efficacious and tolerable among TDT patients when compared to the once-a-day regimen with the same total daily dose. Although there was a significant good results from these studies, there is a need to conduct either muticenter study or randomized control study in a larger number of patients for the better confirmation of the results as all the above studies were conducted in the small number of TDT patients.


Asunto(s)
Deferasirox/uso terapéutico , Quelantes del Hierro/uso terapéutico , Educación del Paciente como Asunto , Talasemia beta/tratamiento farmacológico , Administración Oral , Deferasirox/administración & dosificación , Humanos , Quelantes del Hierro/administración & dosificación
20.
Hemoglobin ; 44(6): 446-450, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33430665

RESUMEN

Thalassemia is a genetic mutation of the α- or ß-globin chains that lead to defective erythropoiesis. This study aimed to collect evidences from all published studies that investigated the clinical effectiveness of calcium channel blockers (CCBs) in conjunction with chelation therapy for reducing iron overload in patients with thalassemia. A systematic search was conducted in PubMed, Institute for Scientific Information (ISI) Web of Science, Scopus, Cochrane Central Register of Controlled Trials, and Virtual Health Library. Original studies reporting the use of CCBs in patients with thalassemia were included for meta-analysis. A total of five randomized studies including 210 patients were included with a follow-up period of 3-12 months. There was no significant difference between amlodipine and control groups in increasing the heart T2* magnetic resonance imaging (MRI) [mean difference (MD) 95% confidence interval (95% CI) = -1.9 (-4.4 to 0.5), p = 0.119] or reducing the liver iron concentration [MD 95% CI = -0.046 (-0.325 to 0.2), p = 0.746]. Although there were no serious adverse events reported in the included trials, further studies are recommended to strengthen our findings.


Asunto(s)
Bloqueadores de los Canales de Calcio/uso terapéutico , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Talasemia beta/complicaciones , Amlodipino/administración & dosificación , Amlodipino/efectos adversos , Amlodipino/uso terapéutico , Biomarcadores , Bloqueadores de los Canales de Calcio/administración & dosificación , Bloqueadores de los Canales de Calcio/efectos adversos , Terapia por Quelación , Quimioterapia Combinada , Humanos , Quelantes del Hierro/administración & dosificación , Quelantes del Hierro/efectos adversos , Sobrecarga de Hierro/diagnóstico , Sobrecarga de Hierro/metabolismo , Imagen por Resonancia Magnética , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento , Talasemia beta/diagnóstico , Talasemia beta/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA