Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Ann Hematol ; 103(6): 2173-2176, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38637332

RESUMEN

A 58-year-old female was found to have hyperferritinemia (Serum ferritin:1683 ng/mL) during work-up for mild normocytic anemia. Transferrin saturation(TSAT) was low-normal. Magnetic resonance imaging (MRI) abdomen showed evidence of hepatic iron deposition. Liver biopsy showed 4 + hepatic iron deposition without any evidence of steatosis or fibrosis. Quantitative liver iron was elevated at 348.3 µmol/g dry liver weight [Reference range(RR): 3-33 µmol/g dry liver weight]. She was presumptively diagnosed with tissue iron overload, cause uncertain. A diagnosis of ferroportin disease (FD) was considered, but the pattern of iron distribution in the liver, mainly within the hepatic parenchyma (rather than in the hepatic Kupffer cells seen in FD), and the presence of anemia (uncommon in FD) made this less likely. She was treated with intermittent phlebotomy for over a decade with poor tolerance due to worsening normocytic to microcytic anemia. A trial of deferasirox was done but it was discontinued after a month due to significant side effects. During the course of treatment, her ferritin level decreased. Over the past 1.5 years, she developed progressively worsening neurocognitive decline. MRI brain showed areas of susceptibility involving basal ganglia, midbrain and cerebellum raising suspicion for metabolic deposition disease. Neuroimaging findings led to testing for serum copper and ceruloplasmin levels which were both found to be severely low. Low serum copper, ceruloplasmin levels and neuroimaging findings led us to consider Wilson disease however prior liver biopsy showing elevated hepatic iron rather than hepatic copper excluded the diagnosis of Wilson disease. After shared decision making, ceruloplasmin gene analysis was not pursued due to patient's preference and prohibitive cost of testing. The diagnosis of aceruloplasminemia was ultimately made. The biochemical triad of hyperferritinemia, low-normal TSAT and microcytic anemia should raise the possibility of aceruloplasminemia. Since neurological manifestations are rare in most inherited iron overload syndromes, neurological symptoms in a patient with tissue iron overload should prompt consideration of aceruloplasminemia as a differential diagnosis.


Asunto(s)
Ceruloplasmina , Trastornos del Metabolismo del Hierro , Imagen por Resonancia Magnética , Humanos , Femenino , Persona de Mediana Edad , Ceruloplasmina/deficiencia , Ceruloplasmina/análisis , Trastornos del Metabolismo del Hierro/diagnóstico , Trastornos del Metabolismo del Hierro/genética , Enfermedades Neurodegenerativas/diagnóstico , Hígado/patología , Hígado/metabolismo , Hígado/diagnóstico por imagen , Ferritinas/sangre , Hierro/metabolismo , Hierro/sangre , Diagnóstico Diferencial , Sobrecarga de Hierro/diagnóstico , Deferasirox/uso terapéutico
2.
Br J Haematol ; 204(5): 2049-2056, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38343073

RESUMEN

Iron overload from repeated transfusions has a negative impact on cardiac function, and iron chelation therapy may help prevent cardiac dysfunction in transfusion-dependent patients with myelodysplastic syndromes (MDS). TELESTO (NCT00940602) was a prospective, placebo-controlled, randomised study to evaluate the iron chelator deferasirox in patients with low- or intermediate-1-risk MDS and iron overload. Echocardiographic parameters were collected at screening and during treatment. Patients receiving deferasirox experienced a significant decrease in the composite risk of hospitalisation for congestive heart failure (CHF) or worsening of cardiac function (HR = 0.23; 95% CI: 0.05, 0.99; nominal p = 0.0322) versus placebo. No significant differences between the arms were found in left ventricular ejection fraction, ventricular diameter and mass or pulmonary artery pressure. The absolute number of events was low, but the enrolled patients were younger than average for patients with MDS, with no serious cardiac comorbidities and a modest cardiovascular risk profile. These results support the effectiveness of deferasirox in preventing cardiac damage caused by iron overload in this patient population. Identification of patients developing CHF is challenging due to the lack of distinctive echocardiographic features. The treatment of iron overload may be important to prevent cardiac dysfunction in these patients, even those with moderate CHF risk.


Asunto(s)
Deferasirox , Quelantes del Hierro , Sobrecarga de Hierro , Síndromes Mielodisplásicos , Humanos , Deferasirox/uso terapéutico , Síndromes Mielodisplásicos/terapia , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/complicaciones , Masculino , Femenino , Quelantes del Hierro/uso terapéutico , Persona de Mediana Edad , Anciano , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/tratamiento farmacológico , Estudios Prospectivos , Benzoatos/uso terapéutico , Benzoatos/efectos adversos , Insuficiencia Cardíaca/etiología , Reacción a la Transfusión/etiología , Ecocardiografía , Adulto , Anciano de 80 o más Años , Triazoles/uso terapéutico , Triazoles/efectos adversos , Transfusión Sanguínea
3.
Artículo en Inglés | MEDLINE | ID: mdl-38423705

RESUMEN

Deferasirox is an iron-chelating drug developed by Novartis company for treatment of diseases accompanied by chronic iron overload; such as ß-thalassemia or sickle cell diseases. Owing to its advantages such as high affinity, specificity and wide therapeutic window, it is considered as first line treatment. The current chapter describes the physicochemical characteristics, mode of action, pharmacokinetics, therapeutic applications and synthetic methods for deferasirox. Moreover, it includes Fourier transform infrared spectrometry (FTIR) and nuclear magnetic resonance spectroscopy (NMR) analysis for its functional groups. In addition, the selected analytical methods are summarized to aid the analysts in their routine analysis of deferasirox.


Asunto(s)
Benzoatos , Sobrecarga de Hierro , Humanos , Deferasirox/farmacología , Deferasirox/uso terapéutico , Benzoatos/farmacología , Benzoatos/uso terapéutico , Benzoatos/metabolismo , Triazoles/uso terapéutico , Triazoles/farmacocinética , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Quelantes del Hierro/metabolismo , Sobrecarga de Hierro/tratamiento farmacológico , Hierro/metabolismo , Hierro/uso terapéutico
4.
Drug Dev Ind Pharm ; 50(3): 223-235, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38305197

RESUMEN

BACKGROUND: Breast cancer (BC) stands as the second-leading cause of mortality among women worldwide. Many chemotherapeutic treatments for BC come with significant adverse effects. Additionally, BC is recognized as one of the most resistant forms of malignancy to treatment. Consequently, there exists a critical need for innovative therapeutic agents that are both highly effective and exhibit reduced toxicity and side effects for patients. Deferasirox (DFX), an iron-chelating drug approved by the FDA for oral use, emerges as a promising contender in the fight against BC proliferation. DFX, primarily administered orally, is utilized to address chronic iron excess resulting from blood transfusions, and it is the inaugural treatment for chronic iron overload syndrome. However, DFX encounters limitations due to its poor water solubility. AIM: This study aimed at incorporating DFX into lipid nanocapsules (DFX-LNCs) followed by investigating the anticancer effect of the DFX nanoform as compared to free DFX in-vitro and on an orthotopic BC mouse model in-vivo. METHODS: The DFX-LNCs was prepared and imaged using TEM and also characterized in terms of particle size (PS), zeta potential (ZP), and polydispersity index (PDI) using DLS. Moreover, drug release, cytotoxicity, and anticancer effect were assessed in-vitro, and in-vivo. RESULTS: The results revealed that DFX-LNCs are more cytotoxic than free DFX with IC50 of 4.417 µg/ml and 16.114 µg/ml, respectively, while the plain LNCs didn't show any cytotoxic effect on the 4T1 cell line (IC50 = 122.797 µg/ml). Besides, the apoptotic effect of DFX-LNCs was more pronounced than that of free DFX, as evidenced by Annexin V/PI staining, increased BAX expression, and decreased expression of BcL-2. Moreover, DFX-LNCs showed a superior antitumor effect in-vivo with potent antioxidant and anti-proliferative effects. CONCLUSION: The newly developed DFX nanoform demonstrated a high potential as a promising therapeutic agent for BC treatment.


Asunto(s)
Neoplasias de la Mama , Sobrecarga de Hierro , Humanos , Femenino , Ratones , Animales , Deferasirox/farmacología , Deferasirox/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Quelantes del Hierro/efectos adversos , Hierro/uso terapéutico , Sobrecarga de Hierro/inducido químicamente , Sobrecarga de Hierro/tratamiento farmacológico
5.
BMJ Open ; 14(2): e077342, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38331857

RESUMEN

INTRODUCTION: Despite the improvement in medical management, many patients with transfusion-dependent ß-thalassaemia die prematurely due to transfusion-related iron overload. As per the current guidelines, the optimal chelation of iron cannot be achieved in many patients, even with two iron chelators at their maximum therapeutic doses. Here, we evaluate the efficacy and safety of triple combination treatment with deferoxamine, deferasirox and deferiprone over dual combination of deferoxamine and deferasirox on iron chelation in patients with transfusion-dependent ß-thalassaemia with very high iron overload. METHODS AND ANALYSIS: This is a single-centre, open-label, randomised, controlled clinical trial conducted at the Adult and Adolescent Thalassaemia Centre of Colombo North Teaching Hospital, Ragama, Sri Lanka. Patients with haematologically and genetically confirmed transfusion-dependent ß-thalassaemia are enrolled and randomised into intervention or control groups. The intervention arm will receive a combination of oral deferasirox, oral deferiprone and subcutaneous deferoxamine for 6 months. The control arm will receive the combination of oral deferasirox and subcutaneous deferoxamine for 6 months. Reduction in iron overload, as measured by a reduction in the serum ferritin after completion of the treatment, will be the primary outcome measure. Reduction in liver and cardiac iron content as measured by T2* MRI and the side effect profile of trial medications are the secondary outcome measures. ETHICS AND DISSEMINATION: Ethical approval for the study has been obtained from the Ethics Committee of the Faculty of Medicine, University of Kelaniya (Ref. P/06/02/2023). The trial results will be disseminated in scientific publications in reputed journals. TRIAL REGISTRATION NUMBER: The trial is registered in the Sri Lanka Clinical Trials Registry (Ref: SLCTR/2023/010).


Asunto(s)
Sobrecarga de Hierro , Talasemia beta , Adulto , Adolescente , Humanos , Deferasirox/uso terapéutico , Deferiprona/uso terapéutico , Deferoxamina/uso terapéutico , Talasemia beta/complicaciones , Talasemia beta/tratamiento farmacológico , Benzoatos/uso terapéutico , Benzoatos/efectos adversos , Triazoles/efectos adversos , Piridonas , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Quelantes del Hierro/efectos adversos , Hierro/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto
6.
PLoS One ; 19(1): e0284267, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38215162

RESUMEN

BACKGROUND AND AIM: Hypozincemia is a prevalent adverse consequence in diabetes mellitus (DM) and ß-Thalassemia patients. We aimed to evaluate the level of serum zinc in ß-thalassemia patients with DM and a risk assessment for hypozincemia. METHODS: The study population included transfusion-dependent thalassemia (TDT) and non-transfusion-dependent thalassemia (NTDT) with overt DM (fasting plasma glucose (FPG) ≥126 mg/dL, and/or 2-h plasma glucose≥200 mg/dL). Serum zinc concentration was measured by the colorimetric method, and the values below 70 µg/dL were defined as hypozincemia. Myocardial and liver T2*-weighted magnetic resonance imaging (MRI T2*, millisecond [ms]) were valued by a free contrast MRI. The demographic, clinical, paraclinical, and laboratory data were also recorded. The data belonged to the period from December 2018 until December 2020. RESULTS: Of 64 diabetic ß-thalassemia patients, 41 cases had zinc data in their medical files (aged 38 ± 9 years, 48.8% female). 78.05% of patients (n = 32) were TDT, and 21.95% were NTDT (n = 9). The mean ± standard deviation of zinc level was 110.2 ± 127.6 µg/dL. The prevalence of hypozincemia was 9.76%, 95% confidence interval [CI] 0.27 to 19.24 (four cases). After controlling age, the odds of hypozincemia for using deferasirox (DFX) was 8.77, 95% CI 0.60 to 127.1. In ß-thalassemia patients, the age-adjusted risk of hypozincemia was calculated at 15.85, 95% CI 0.47 to 529.3 for hepatitis C. The adjusted risk of hypozincemia based on age for antacid use was 6.34, 95% CI 0.39 to 102.7. CONCLUSION: In light of this study, as well as hepatitis C, using DFX and antacids is associated with a high risk of hypozincemia amid diabetic ß-thalassemia cases. However, upward bias should be taken into consideration.


Asunto(s)
Diabetes Mellitus , Hepatitis C , Sobrecarga de Hierro , Talasemia , Talasemia beta , Humanos , Femenino , Masculino , Talasemia beta/complicaciones , Talasemia beta/tratamiento farmacológico , Deferasirox/uso terapéutico , Sobrecarga de Hierro/complicaciones , Glucemia , Factores de Riesgo , Talasemia/epidemiología , Hepatitis C/complicaciones , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/inducido químicamente , Zinc , Quelantes del Hierro/uso terapéutico
7.
Auris Nasus Larynx ; 51(2): 271-275, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37903661

RESUMEN

OBJECTIVE: The role of iron chelation in causing hearing loss (HL) is still unclear. The present study assessed the prevalence of HL among transfusion-dependent thalassemia (TDT) patients who underwent audiological follow-up over a 20-year period. METHODS: We retrospectively analyzed clinical records and audiological tests from January 1990 (T0) to December 2022 (T22) of a group of TDT patients who received iron chelation therapy with deferoxamine (DFO), deferiprone (DFP) or deferasirox (DFX), in monotherapy or as part of combination therapy. RESULTS: A total of 42 adult TDT patients (18 male, 24 female; age range: 41-55 years; mean age: 49.2 ± 3.7 years) were included in the study. At the T22 assessment, the overall prevalence of sensorineural HL was 23.8 % (10/42). When patients were stratified into two groups, with and without ototoxicity, no differences were observed for sex, age, BMI, creatinine level, pre-transfusional hemoglobin, start of transfusions, cardiac or hepatic T2 MRI; only ferritin serum values and duration of chelation were significantly higher (p = 0.02 and p = 0.01, respectively) in patients with hearing impairment in comparison to those with normal hearing. CONCLUSION: This study with long-term follow-up suggests that iron chelation therapy might induce ototoxicity; therefore, a long and accurate audiological follow-up should be performed in TDT patients.


Asunto(s)
Sobrecarga de Hierro , Ototoxicidad , Talasemia beta , Adulto , Humanos , Masculino , Femenino , Persona de Mediana Edad , Talasemia beta/complicaciones , Talasemia beta/tratamiento farmacológico , Talasemia beta/epidemiología , Deferasirox/uso terapéutico , Deferiprona/uso terapéutico , Deferoxamina/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/epidemiología , Sobrecarga de Hierro/etiología , Estudios de Seguimiento , Estudios Retrospectivos , Ototoxicidad/complicaciones , Ototoxicidad/tratamiento farmacológico , Benzoatos/uso terapéutico , Triazoles/uso terapéutico , Piridonas/uso terapéutico , Quelantes del Hierro/uso terapéutico , Hierro/uso terapéutico , Audición
8.
J Am Heart Assoc ; 13(1): e031219, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38158218

RESUMEN

BACKGROUND: Ferroptosis, an iron-dependent form of regulated cell death, is a major cell death mode in myocardial ischemia reperfusion (I/R) injury, along with mitochondrial permeability transition-driven necrosis, which is inhibited by cyclosporine A (CsA). However, therapeutics targeting ferroptosis during myocardial I/R injury have not yet been developed. Hence, we aimed to investigate the therapeutic efficacy of deferasirox, an iron chelator, against hypoxia/reoxygenation-induced ferroptosis in cultured cardiomyocytes and myocardial I/R injury. METHODS AND RESULTS: The effects of deferasirox on hypoxia/reoxygenation-induced iron overload in the endoplasmic reticulum, lipid peroxidation, and ferroptosis were examined in cultured cardiomyocytes. In a mouse model of I/R injury, the infarct size and adverse cardiac remodeling were examined after treatment with deferasirox, CsA, or both in combination. Deferasirox suppressed hypoxia- or hypoxia/reoxygenation-induced iron overload in the endoplasmic reticulum, lipid peroxidation, and ferroptosis in cultured cardiomyocytes. Deferasirox treatment reduced iron levels in the endoplasmic reticulum and prevented increases in lipid peroxidation and ferroptosis in the I/R-injured myocardium 24 hours after I/R. Deferasirox and CsA independently reduced the infarct size after I/R injury to a similar degree, and combination therapy with deferasirox and CsA synergistically reduced the infarct size (infarct area/area at risk; control treatment: 64±2%; deferasirox treatment: 48±3%; CsA treatment: 48±4%; deferasirox+CsA treatment: 37±3%), thereby ameliorating adverse cardiac remodeling on day 14 after I/R. CONCLUSIONS: Combination therapy with deferasirox and CsA may be a clinically feasible and effective therapeutic approach for limiting I/R injury and ameliorating adverse cardiac remodeling after myocardial infarction.


Asunto(s)
Ferroptosis , Sobrecarga de Hierro , Infarto del Miocardio , Isquemia Miocárdica , Daño por Reperfusión Miocárdica , Daño por Reperfusión , Ratones , Animales , Ciclosporina/farmacología , Daño por Reperfusión Miocárdica/metabolismo , Deferasirox/farmacología , Deferasirox/metabolismo , Deferasirox/uso terapéutico , Remodelación Ventricular , Miocitos Cardíacos/metabolismo , Infarto del Miocardio/metabolismo , Daño por Reperfusión/metabolismo , Hierro/metabolismo , Hipoxia/metabolismo , Sobrecarga de Hierro/metabolismo , Isquemia Miocárdica/metabolismo
9.
Rev Alerg Mex ; 70(4): 159-162, 2023 Sep.
Artículo en Español | MEDLINE | ID: mdl-37933925

RESUMEN

Background: Deferasirox is an active iron chelator, used in the treatment of iron overload such as hemochromatosis. Up to 28% may present adverse reactions to said drug. A desensitization protocol for this drug may be useful when there are no other therapeutic options. Case report: A 52-year-old female with a diagnosis of hemochromatosis who began treatment with phlebotomy, poor response and tolerance, so it was decided to treat with deferasirox 500 mg daily, presenting symptoms of urticaria and angioedema on the third dose. Hospitalization was decided for a desensitization protocol with an initial dose of 0.6mg with a gradual increase in the dose, reaching a maintenance dose of 500 mg per day on the third day. Conclusions: The rapid desensitization protocol for Deferasirox is useful when there is no response or therapeutic alternative.


Antecedentes: Deferasirox es un quelante de hierro activo, indicado en el tratamiento de pacientes con hemocromatosis; sin embargo, se ha informado que el 28% de los casos puede tener reacciones adversas al fármaco. El protocolo de desensibilización para deferasirox puede ser útil cuando no se dispone de opciones terapéuticas adicionales. Reporte de caso: Paciente femenina de 52 años, con diagnóstico de hemocromatosis, quien luego de practicarle una flebotomía se observó poca respuesta y tolerancia al tratamiento, por lo que se decidió indicar deferasirox (500 mg/día), manifestando un cuadro de urticaria y angioedema en la tercera toma. Se decidió hospitalizarla para implementar el protocolo de desensibilización con una dosis inicial de 0.6 mg, con incremento gradual hasta llegar, al tercer día, a una dosis de mantenimiento de 500 mg/día. Conclusiones: El protocolo de desensibilización rápida con deferasirox es útil cuando no se obtiene respuesta satisfactoria con la flebotomía o no se dispone opciones de tratamiento alternativas.


Asunto(s)
Deferasirox , Hemocromatosis , Quelantes del Hierro , Femenino , Humanos , Persona de Mediana Edad , Deferasirox/uso terapéutico , Hemocromatosis/tratamiento farmacológico , Quelantes del Hierro/uso terapéutico
10.
Complement Ther Clin Pract ; 53: 101804, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37832335

RESUMEN

BACKGROUND AND PURPOSE: Iron overload in the body is associated with serious and irreversible tissue damage. This study aimed to investigate the iron-chelating, antioxidant, anti-inflammatory, and hepatoprotective activities of grape seed extract (GSE) supplement as well as its safety in ß-thalassemia major (ß-TM) pediatric patients receiving deferasirox as a standard iron-chelation therapy. MATERIALS AND METHODS: The children were randomly allocated to either GSE group (n = 30) or control group (n = 30) to receive GSE (100 mg/day) or placebo capsules, respectively, for 4 weeks. The serum levels of iron, ferritin, total iron-binding capacity (TIBC), alanine transaminase (ALT), aspartate aminotransferase (AST), tumor necrosis factor alpha (TNF-α), high-sensitivity C-reactive protein (hs-CRP), malondialdehyde (MDA), and glutathione (GSH) as well as superoxide dismutase (SOD) activity and hemoglobin (Hb) concentration were measured pre-and post-intervention. RESULTS: GSE supplement significantly attenuated the serum levels of iron (p = 0.030), ferritin (p = 0.017), ALT (p = 0.000), AST (p = 0.000), TNF-α (p = 0.000), and hs-CRP (p = 0.001). The TIBC level (p = 0.020) significantly enhanced in the GSE group compared with the placebo group. Moreover, GSE supplement remarkably improved the oxidative stress markers, MDA (p = 0.000) and GSH (p = 0.001). The changes in the SOD activity (p = 0.590) and Hb concentration (p = 0.670) were not statistically different between the groups. CONCLUSION: GSE supplement possesses several health beneficial influences on children with ß-TM by alleviating iron burden, oxidative stress, inflammation, and liver dysfunction.


Asunto(s)
Extracto de Semillas de Uva , Sobrecarga de Hierro , Hepatopatías , Talasemia beta , Niño , Humanos , Talasemia beta/tratamiento farmacológico , Talasemia beta/complicaciones , Proteína C-Reactiva , Deferasirox/uso terapéutico , Ferritinas/metabolismo , Extracto de Semillas de Uva/farmacología , Extracto de Semillas de Uva/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/complicaciones , Hierro/metabolismo , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/metabolismo , Hepatopatías/complicaciones , Estrés Oxidativo , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa
11.
Ann N Y Acad Sci ; 1529(1): 33-41, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37594980

RESUMEN

Combination chelation therapies are considered in transfusion-dependent thalassemia patients for whom monotherapy regimens have failed to achieve iron balance or intensification of iron chelation therapy is required for the rapid reduction of excess iron to avoid permanent organ damage. Combination chelation may provide a more flexible approach for individualizing chelation therapy, thereby improving tolerability, adherence, and quality of life. In principle, iron chelators can be combined with an infinite number of dosing regimens; these involve simultaneous or sequential exposure to the chelators on the same day or alternating the drugs on different days. Clinical studies have established the safety and efficacy of chelation combinations. However, real-life data with combination therapies indicate the significance of compliance for a meaningful reduction in iron overload compared to monotherapies.


Asunto(s)
Terapia por Quelación , Sobrecarga de Hierro , Humanos , Deferasirox/uso terapéutico , Deferoxamina/uso terapéutico , Deferiprona/uso terapéutico , Calidad de Vida , Benzoatos/efectos adversos , Triazoles , Piridonas , Quelantes del Hierro/uso terapéutico , Quelantes del Hierro/efectos adversos , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/inducido químicamente , Hierro , Quimioterapia Combinada
12.
Hemodial Int ; 27(3): 270-277, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36994679

RESUMEN

INTRODUCTION: Hemosiderosis of chronic dialysis has always been a frequent phenomenon in dialysis; formerly related to blood transfusions before the advent of Erythropoiesis Stimulating Agents (ESA), it is currently in connection with the use of massive doses of injectable iron, to ensure the full therapeutic efficacy of ESA. Few studies have looked at the therapeutic aspect of iron chelators in the dialysis population. METHODS: We followed 31 dialysis patients treated for secondary hemosiderosis with deferasirox (DFX) at the dose 10 mg/kg/day, by hepatic MRI from September 2017 to September 2021, in order to evaluate the efficacy of iron chelators on the reduction of liver iron concentration (LIC). The diagnosis of hemosiderosis was carried for a value of the LIC > 50 µmol/g of dry liver. RESULTS: Chelation resulted in a significant reduction in liver iron burden as measured by liver MRI: (201.4 ± 179.9 vs. 122.6 ± 154.3 µmol/g liver) (p = 0.000) and in mean ferritin level: (2058.8 ± 2004.9 vs. 644.2 ± 456.6 ng/mL) (p = 0.002). A gain of 1.1 g/dL in mean hemoglobin level: (10.5 ± 1.6 vs. 11.6 ± 2.0 g/dL) (p = 0.006). A significant increase in mean albumin level: (43 ± 5.5 to 46.2 ± 6.1 g/L) (p = 0.04). The therapeutic response was clearly influenced by the cause of overload, longer in polytransfused patients (p = 0.023) and the degree of overload assessed by MRI (p = 0.003) and ferritin level (p = 0.04). CONCLUSION: DFX, prescribed at a dose of 10 mg/kg/day, resulted in a significant reduction in hepatic iron burden as measured by liver MRI and ferritin. The therapeutic response was clearly influenced by blood transfusions and the degree of iron overload.


Asunto(s)
Hemosiderosis , Sobrecarga de Hierro , Humanos , Deferasirox/uso terapéutico , Ferritinas/uso terapéutico , Hemosiderosis/etiología , Hemosiderosis/complicaciones , Hierro/uso terapéutico , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Hígado/diagnóstico por imagen , Hígado/metabolismo , Hígado/patología , Imagen por Resonancia Magnética , Diálisis Renal/efectos adversos
13.
Expert Rev Hematol ; 16(2): 81-94, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36755516

RESUMEN

INTRODUCTION: Regular blood transfusions in patients with thalassemia syndromes can cause iron overload resulting in complications including cirrhosis, heart problems, or endocrine abnormalities. To prevent iron overload toxicity in these patients, three iron chelators are currently FDA-approved for use: deferoxamine, deferasirox, and deferiprone. In the United States, deferiprone has been approved for three times daily dosing since 2011 and has recently gained approval for twice-daily administration. AREAS COVERED: A PubMed literature search was performed with the keywords 'deferiprone' and 'thalassemia.' Relevant original research studying deferiprone's effects on transfusional iron overload in patients with thalassemia syndromes was included. Exclusion criteria included case reports and review papers. Deferiprone is effective at reducing serum ferritin levels in patients with iron overload. Twice-daily administration provides a similar level of iron chelation as three times daily dosing with a comparable side effect profile and increased patient acceptability. EXPERT OPINION: New studies are highlighting deferiprone's potential for combination therapy with either deferoxamine or deferasirox to improve iron chelation. Deferiprone's ability to significantly decrease cardiac and liver iron content can be utilized in other transfusion-dependent hematologic conditions, as evidenced by its recent approval for use in the United States for sickle cell disease or other anemias.


Asunto(s)
Sobrecarga de Hierro , Talasemia , Humanos , Deferasirox/uso terapéutico , Deferoxamina/uso terapéutico , Síndrome , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Quelantes del Hierro/uso terapéutico , Talasemia/complicaciones , Talasemia/terapia , Transfusión Sanguínea , Piridonas/efectos adversos , Benzoatos
14.
Eur J Haematol ; 110(5): 490-497, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36708354

RESUMEN

Iron overload is a pathological condition resulting from a congenital impairment of its regulation, increased intestinal iron absorption secondary to bone marrow erythroid hyperplasia, or a chronic transfusional regimen. In normal conditions, intracellular and systemic mechanisms contribute to maintaining iron balance. When this complex homeostatic mechanism fails, an iron overload could be present. Detecting an iron overload is not easy. The gold standard remains the liver biopsy, even if it is invasive and dangerous. Identifying iron using noninvasive techniques allowed a better understanding of the rate of iron overload in different organs, with a low risk for the patient. Estimating serum ferritin (mg/L) is the easiest and, consequently, the most employed diagnostic tool for assessing body iron stores, even if it could be a not specific method. The most common hematological causes of iron overload are myelodysplastic syndromes, sickle cell disease, and thalassemia. In all of these conditions, three drugs have been approved for the treatment of iron overload: deferiprone, deferoxamine, and deferasirox. These chelators have been demonstrated to help lower tissue iron levels and prevent iron overload complications, improving event-free survival (EFS). Nowadays, the decision to start chelation and which chelator to choose remains the joint decision of the clinician and patient.


Asunto(s)
Terapia por Quelación , Sobrecarga de Hierro , Humanos , Terapia por Quelación/efectos adversos , Quelantes del Hierro/uso terapéutico , Deferasirox/uso terapéutico , Deferiprona/uso terapéutico , Deferoxamina/uso terapéutico , Piridonas/uso terapéutico , Benzoatos/uso terapéutico , Triazoles , Sobrecarga de Hierro/diagnóstico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Hierro
15.
J Inorg Biochem ; 241: 112131, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36706491

RESUMEN

Capillary electrophoresis was used to estimate the solvolytic dissociation rate (kd) of metal complexes of deferasirox (DFX, H3L), a drug used to treat iron overload. Inert CoIIIL23- did not dissociate. The estimated kd value for FeIIIL23- was (2.7 ± 0.3) × 10-4 s-1 (298 K, pH 7.4). The kd values of other complexes (AlIIIL23-, NiIIL24-, and MnIIL-) were in the range 10-3-10-4 s-1. In contrast, ZnIIL- and CuIIL- were too labile to allow kd estimation. The fact that the half-life of FeIIIL23- (43.3 min) is shorter than the blood half-life of DFX (8-16 h) implies that the blood concentration of DFX should be high enough to prevent dissociation of FeIIIL23-. The possibility of a safer iron-chelation therapy that avoids excretion of other essential metal ions such as ZnII is discussed, highlighting the importance of selectivity in terms of kinetic stability.


Asunto(s)
Sobrecarga de Hierro , Hierro , Humanos , Deferasirox/uso terapéutico , Terapia por Quelación , Quelantes del Hierro , Electroforesis Capilar , Benzoatos
16.
Transplant Cell Ther ; 29(1): 42.e1-42.e6, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36241148

RESUMEN

During conditioning chemotherapy prior to allogeneic haematopoietic stem cell transplantation (HSCT), non-transferrin-bound iron and its chelatable form, labile plasma iron (LPI), regularly appear in the blood of patients at high levels of transferrin saturation (TfS). As these free iron species potentially favor infection and mediate transplantation-associated toxicities, chelation therapy could be an approach to improve outcome after transplantation. However, data addressing iron chelation in the immediate peritransplantation period are sparse. In this study, we investigated the influence of iron chelation with deferasirox during conditioning chemotherapy on the appearance of LPI, the incidence of infection and toxicities, and the tolerability of this treatment in the peritransplantation period. We conducted this single-center prospective observational study in 25 adults with iron overload (serum ferritin >1000 µg/L) undergoing allogeneic HSCT after myeloablative busulfan-based conditioning chemotherapy. Patients received iron chelation with deferasirox (14 mg/kg) from the start of conditioning until day 3 post-transplantation. Iron parameters, including LPI, were obtained at the chelator's trough level daily until day 0 and then on days 4, 7, and 14. Data on infection (bacteremia or invasive fungal disease) and toxicity, as well as the tolerability of deferasirox, were collected until the end of the follow-up period on day 28. Data were analyzed descriptively. TfS levels exceeded 70% in median on 6 days (range, 4 to 10 days) and in 63.6% (range, 36.4% to 90.9%) of the samples per patient, although in 19 of 25 patients (76%), no elevated LPI values were detected during the intake of deferasirox despite high TfS levels. Only 6 patients (24%) showed mildly increased LPI values (≤0.5 units) during the intake of deferasirox, 3 of whom had presented with elevated LPI values before the start of conditioning. Deferasirox was well tolerated, and no aggravation of toxicities was observed. Infection occurred in 5 patients (20%), including 3 of the 6 patients with elevated LPI values despite chelation therapy. In the present study, we demonstrate that iron chelation with deferasirox safely suppresses the appearance of LPI and might decrease the incidence of infection, whereas the impact on transplantation-associated toxicities remains to be elucidated.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Hierro , Adulto , Humanos , Deferasirox/uso terapéutico , Ferritinas , Quelantes del Hierro/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/efectos adversos
17.
Transfus Clin Biol ; 30(1): 69-74, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35878782

RESUMEN

OBJECTIVES: Iron overload is a common complication experienced by transfusion-dependent children with hemoglobin disorders. Chelators such as deferasirox (DFX) and deferiprone (DFP) are effective in overcoming this problem. We conducted this systematic review and meta-analysis to evaluate the effectiveness of DFX compared to DFP in treating iron overload amongst pediatric patients with hemoglobin disorders. MATERIAL AND METHODS: PubMed and Cochrane Central were searched from their inception until Dec 21 2021, for randomized clinical trials (RCTs) and observational studies, which assessed the efficacy of DFX compared to DFP in the treatment of inherited hemoglobin disorders. The outcomes of interest included myocardial iron concentration (MRI T2*) at the end of the trial and change in mean serum ferritin (SF) levels at the 6 and 12 months mark. Weighted mean differences (WMDs) with their corresponding 95% confidence intervals (CIs) were calculated for continuous outcomes using random effects model. RESULTS: A total of 5 studies comprising 607 children were included. The results of our analysis revealed no significant difference between DFX and DFP in MRI T2* at the end of treatment (WMD: -0.92; 95% CI [-3.35, 1.52]; p = 0.46; I2 = 0). Moreover, there has been no significant difference noted in SF levels at both 6 months (WMD: 97.31; 95% CI [-236.16, 430.77]; p = 0.57; I2 = 0) and 12 months (WMD: 46.99; 95% CI [-191.42, 285.40]; p = 0.70; I2 = 0) respectively. CONCLUSION: Our analysis shows no significant difference between the efficacy of DFX and DFP in the management of iron overload in children with inherited blood disorders. Future large-scale clinical trials are required to further validate our results.


Asunto(s)
Hemoglobinopatías , Sobrecarga de Hierro , Talasemia beta , Humanos , Niño , Hierro/uso terapéutico , Hierro/metabolismo , Deferasirox/uso terapéutico , Deferiprona/uso terapéutico , Quelantes del Hierro/uso terapéutico , Benzoatos/uso terapéutico , Triazoles/uso terapéutico , Piridonas/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Talasemia beta/complicaciones , Talasemia beta/tratamiento farmacológico , Hemoglobinopatías/complicaciones , Hemoglobinopatías/tratamiento farmacológico , Ferritinas
18.
J Int Med Res ; 50(12): 3000605221143290, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36562113

RESUMEN

OBJECTIVES: To examine the efficacy of deferasirox (DFX) by comparison with deferoxamine (DFO) in managing iron overload in patients with sickle cell anaemia (SCA). METHODS: Online databases were systematically searched for studies published from January 2007 to July 2022 that had investigated the efficacy of DFX compared with DFO in managing iron overload in patients with SCA. RESULTS: Of the 316 articles identified, three randomized clinical trials met the inclusion criteria. Meta-analysis of liver tissue iron concentration (LIC) showed that iron overload was not significantly higher in the DFX group compared with DFO group (WMD, -1.61 mg Fe/g dw (95% CI -4.42 to 1.21). However, iron overload as measured by serum ferritin was significantly lower in DFO compared with DFX group (WMD, 278.13 µg/l (95% CI 36.69 to 519.57). Although meta-analysis was not performed on myocardial iron concentration due to incomplete data, the original report found no significant difference between DFX and DFO. CONCLUSION: While limited by the number of studies included in this meta-analysis, overall, the results tend to show that DFX was as effective as DFO in managing iron overload in patients with SCA.


Asunto(s)
Anemia de Células Falciformes , Sobrecarga de Hierro , Humanos , Deferasirox/uso terapéutico , Deferoxamina/uso terapéutico , Quelantes del Hierro/uso terapéutico , Benzoatos/uso terapéutico , Triazoles/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Hierro , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/tratamiento farmacológico
19.
J Med Chem ; 65(21): 14692-14700, 2022 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-36353870

RESUMEN

DNA damage repair is considered to be an important mechanism of cisplatin resistance, and the roles of iron homeostasis in action mechanisms of cisplatin have not been studied yet. Herein, a Pt(IV) prodrug (DFX-Pt) integrating cisplatin and the clinical oral iron-chelating agent deferasirox (DFX) is found to be highly active toward cisplatin-insensitive triple-negative breast cancer cells both in vitro and in vivo. RNA-sequencing shows that DFX-Pt can downregulate genes related to the double-strand break (DSB) damage pathway significantly. DFX-Pt can reduce cellular free iron, regulate the expression of the RNA demethylase, and elevate the levels of RNA N6-methyladenosine (m6A), which degrades the DSB-related genes in an m6A-dependent manner. In all, we first reveal the roles of RNA modification in mechanisms of combating DNA damage repair and show that the combination of iron homeostasis intervention may bring new treatment regimens for cisplatin resistance.


Asunto(s)
Profármacos , Humanos , Profármacos/farmacología , Deferasirox/uso terapéutico , Cisplatino/farmacología , Metilación , ARN , Quelantes del Hierro/uso terapéutico , Hierro , Daño del ADN
20.
Hemoglobin ; 46(2): 114-117, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36069257

RESUMEN

Red blood cell (RBC) transfusions have been established as one of the primary therapies in treating sickle cell anemia. However, they are not free of side effects, with overloading the body with iron being one of the most important. Iron chelation therapy greatly reduces the iron load of the body. In addition, hydroxyurea (HU), an oral chemotherapeutic drug also has a significant role in the treatment of the disease with beneficial effects on many of the clinical problems that arise, mainly in reducing painful crises. The aim of this study was to investigate the effect of synergistic transfusion therapy and HU on the response to deferasirox (DFX) chelation therapy. Eighteen patients with sickle cell disease were divided into two groups based on their treatment, either with simple transfusions and DFX or with a combination of transfusion therapy, DFX and HU, and were evaluated with magnetic resonance imaging (MRI) for liver iron concentration (LIC) and biochemistry. All patients completed the study. The results of the study showed improvement in serum ferritin (FER) levels and LIC after 12 months of therapy in both groups, especially in the group receiving the combination therapy with HU. In addition, there was a noteworthy improvement in serum glutamic-oxaloacetic transaminase (SGOT), serum glutamic pyruvic transaminase (SGPT) and lactate dehydrogenase (LDH) levels with serum creatinine (Cr) levels remaining stable during the study in both groups. Hydroxyurea, when combined with iron chelators such as DFX, provides an additional benefit of iron chelation in patients receiving chronic transfusion therapy.


Asunto(s)
Anemia de Células Falciformes , Quelantes del Hierro , Sobrecarga de Hierro , Alanina Transaminasa/uso terapéutico , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/terapia , Aspartato Aminotransferasas/uso terapéutico , Terapia por Quelación , Creatinina/uso terapéutico , Deferasirox/uso terapéutico , Ferritinas , Humanos , Hidroxiurea/uso terapéutico , Hierro , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/etiología , Lactato Deshidrogenasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...