Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharm ; 18(2): 593-609, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32926630

RESUMEN

Deferoxamine mesylate (DFO) is an FDA-approved, hexadentate iron chelator routinely used to alleviate systemic iron burden in thalassemia major and sickle cell patients. Iron accumulation in these disease states results from the repeated blood transfusions required to manage these conditions. Iron accumulation has also been implicated in the pathogenesis of Alzheimer's disease (AD), Parkinson's disease (PD), and secondary injury following intracerebral hemorrhage (ICH). Chelation of brain iron is thus a promising therapeutic strategy for improving behavioral outcomes and slowing neurodegeneration in the aforementioned disease states, though the effectiveness of DFO treatment is limited on several accounts. Systemically administered DFO results in nonspecific toxicity at high doses, and the drug's short half-life leads to low patient compliance. Mixed reports of DFO's ability to cross the blood-brain barrier (BBB) also appear in literature. These limitations necessitate novel DFO formulations prior to the drug's widespread use in managing neurodegeneration. Herein, we discuss the various dosing regimens and formulations employed in intranasal (IN) or systemic DFO treatment, as well as the physiological and behavioral outcomes observed in animal models of AD, PD, and ICH. The clinical progress of chelation therapy with DFO in managing neurodegeneration is also evaluated. Finally, the elimination of intranasally administered particles via the glymphatic system and efflux transporters is discussed. Abundant preclinical evidence suggests that intranasal DFO treatment improves memory retention and behavioral outcome in rodent models of AD, PD, and ICH. Several other biochemical and physiological metrics, such as tau phosphorylation, the survival of tyrosine hydroxylase-positive neurons, and infarct volume, are also positively affected by intranasal DFO treatment. However, dosing regimens are inconsistent across studies, and little is known about brain DFO concentration following treatment. Systemic DFO treatment yields similar results, and some complex formulations have been developed to improve permeability across the BBB. However, despite the success in preclinical models, clinical translation is limited with most clinical evidence investigating DFO treatment in ICH patients, where high-dose treatment has proven dangerous and dosing regimens are not consistent across studies. DFO is a strong drug candidate for managing neurodegeneration in the aging population, but before it can be routinely implemented as a therapeutic agent, dosing regimens must be standardized, and brain DFO content following drug administration must be understood and controlled via novel formulations.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Hemorragia Cerebral/tratamiento farmacológico , Deferoxamina/administración & dosificación , Portadores de Fármacos/química , Enfermedad de Parkinson/tratamiento farmacológico , Sideróforos/administración & dosificación , Administración Intranasal , Enfermedad de Alzheimer/patología , Animales , Disponibilidad Biológica , Barrera Hematoencefálica/metabolismo , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/patología , Deferoxamina/farmacocinética , Modelos Animales de Enfermedad , Semivida , Humanos , Inyecciones Intramusculares , Inyecciones Intraventriculares , Inyecciones Espinales , Inyecciones Subcutáneas , Hierro/metabolismo , Cumplimiento de la Medicación , Nanopartículas/química , Mucosa Nasal/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Enfermedad de Parkinson/patología , Permeabilidad , Sideróforos/farmacocinética , Distribución Tisular
2.
Wound Repair Regen ; 26(3): 300-305, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-30152571

RESUMEN

Chronic wounds are a significant medical and economic problem worldwide. Individuals over the age of 65 are particularly vulnerable to pressure ulcers and impaired wound healing. With this demographic growing rapidly, there is a need for effective treatments. We have previously demonstrated that defective hypoxia signaling through destabilization of the master hypoxia-inducible factor 1α (HIF-1α) underlies impairments in both aging and diabetic wound healing. To stabilize HIF-1α, we developed a transdermal delivery system of the Food and Drug Administration-approved small molecule deferoxamine (DFO) and found that transdermal DFO could both prevent and treat ulcers in diabetic mice. Here, we demonstrate that transdermal DFO can similarly prevent pressure ulcers and normalize aged wound healing. Enhanced wound healing by DFO is brought about by stabilization of HIF-1α and improvements in neovascularization. Transdermal DFO can be rapidly translated into the clinic and may represent a new approach to prevent and treat pressure ulcers in aged patients.


Asunto(s)
Deferoxamina/farmacología , Úlcera por Presión/prevención & control , Sideróforos/farmacología , Cicatrización de Heridas/efectos de los fármacos , Administración Cutánea , Animales , Deferoxamina/administración & dosificación , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Úlcera por Presión/fisiopatología , Sideróforos/administración & dosificación , Cicatrización de Heridas/fisiología
3.
BMC Ophthalmol ; 18(1): 246, 2018 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-30208862

RESUMEN

BACKGROUND: Deferoxamine (DFO) is one of the most commonly used chelation treatments for transfusional hemosiderosis. Pattern dystrophies constitute a distinct entity of retinal disorders that has been occasionally identified in association with deferoxamine. CASE PRESENTATION: We report two cases of bilateral macular pattern dystrophy in transfusion dependent patients undergoing chronic chelation therapy with deferoxamine due to thalassemias. Our patients were evaluated with multimodal imaging and the results are presented. Both patients had normal cone and rod responses in the full-field electroretinogram and continued the prescribed chelation therapy, after hematology consult. The patients were followed up every 3 months for 2 and 4 years respectively for possible deterioration. Their best corrected visual acuity remained stable with no anatomic change on Optical Coherence Tomography findings. CONCLUSION: Multimodal imaging of our patients allowed a better evaluation and possibly earlier detection of the DFO-related changes. Screening and close follow up of patients under chronic chelating therapy is important in order to promptly diagnose and manage possible toxicity either with discontinuation of the offending agent or dose modification.


Asunto(s)
Deferoxamina/efectos adversos , Retina/diagnóstico por imagen , Degeneración Retiniana/inducido químicamente , Talasemia/tratamiento farmacológico , Deferoxamina/administración & dosificación , Electrorretinografía , Femenino , Humanos , Infusiones Subcutáneas/efectos adversos , Persona de Mediana Edad , Degeneración Retiniana/diagnóstico , Sideróforos/administración & dosificación , Sideróforos/efectos adversos , Tomografía de Coherencia Óptica
4.
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
5.
Retina ; 37(6): 1168-1175, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27652916

RESUMEN

PURPOSE: To evaluate possible benefits of using electrophysiological investigations for detecting retinal and visual pathway changes and correlating them with chelation modality in children with thalassemia. METHODS: This study included 60 patients on single oral iron chelator (deferasirox) (Group 1), 60 on deferoxamine chelator (Group 2), and 60 controls (Group 3). Participants underwent full ophthalmologic examination, pattern visual-evoked potential, pattern electroretinogram, and multifocal electroretinogram. RESULTS: Fundus showed no abnormalities. Multifocal electroretinogram mean P1 amplitude showed statistically significant differences in all 5 rings, amplitudes being significantly lower in Groups 1 and 2 than Group 3; moreover, significantly lower in Group 2 than Group 1. There was a statistically significant difference between groups regarding P50 wave latency and N35-P50 of pattern electroretinogram amplitude (P < 0.001 and P < 0.001, respectively). However, there were no statistically significant differences between groups regarding N95 wave of pattern electroretinogram and pattern visual-evoked potential waves' amplitude and latency. Multiple regression analyses illustrated that chelator was the most important determinant for multifocal electroretinogram and P50 parameters. CONCLUSION: Preclinical electrophysiologic changes existed in thalassemics, more obvious in those on deferoxamine. Electrophysiologic studies analysis denotes an early toxic macular insult rather than optic nerve affection. Thus, regular follow-up using multifocal electroretinogram and pattern electroretinogram is recommended.


Asunto(s)
Benzoatos/administración & dosificación , Deferoxamina/administración & dosificación , Electrorretinografía , Potenciales Evocados Visuales , Nervio Óptico/fisiopatología , Enfermedades de la Retina/tratamiento farmacológico , Talasemia/fisiopatología , Triazoles/administración & dosificación , Administración Oral , Adolescente , Estudios Transversales , Deferasirox , Relación Dosis-Respuesta a Droga , Oftalmopatías , Femenino , Humanos , Quelantes del Hierro/administración & dosificación , Masculino , Nervio Óptico/diagnóstico por imagen , Retina/efectos de los fármacos , Retina/patología , Retina/fisiopatología , Enfermedades de la Retina/diagnóstico , Enfermedades de la Retina/etiología , Estudios Retrospectivos , Sideróforos/administración & dosificación , Talasemia/complicaciones , Talasemia/tratamiento farmacológico , Resultado del Tratamiento , Agudeza Visual
6.
Int J Mol Sci ; 18(12)2017 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-29261151

RESUMEN

Iron overload (IOL) due to increased intestinal iron absorption constitutes a major clinical problem in patients with non-transfusion-dependent thalassemia (NTDT), which is a cumulative process with advancing age. Current models for iron metabolism in patients with NTDT suggest that suppression of serum hepcidin leads to an increase in iron absorption and subsequent release of iron from the reticuloendothelial system, leading to depletion of macrophage iron, relatively low levels of serum ferritin, and liver iron loading. The consequences of IOL in patients with NTDT are multiple and multifactorial. Accurate and reliable methods of diagnosis and monitoring of body iron levels are essential, and the method of choice for measuring iron accumulation will depend on the patient's needs and on the available facilities. Iron chelation therapy (ICT) remains the backbone of NTDT management and is one of the most effective and practical ways of decreasing morbidity and mortality. The aim of this review is to describe the mechanism of IOL in NTDT, and the clinical complications that can develop as a result, in addition to the current and future therapeutic options available for the management of IOL in NTDT.


Asunto(s)
Sobrecarga de Hierro/tratamiento farmacológico , Sideróforos/uso terapéutico , Talasemia/tratamiento farmacológico , Ensayos Clínicos como Asunto , Humanos , Hierro/metabolismo , Sobrecarga de Hierro/diagnóstico , Sobrecarga de Hierro/etiología , Sideróforos/administración & dosificación , Sideróforos/efectos adversos , Sideróforos/farmacología , Talasemia/complicaciones
7.
Ann Plast Surg ; 77(5): 560-568, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26808734

RESUMEN

BACKGROUND: Autogenous bone grafts are still the criterion standard treatment option in critical-size bone defect reconstructions, and many therapies can affect its incorporation. In this study, it was aimed to research the effects of desferroxamine (DFO) application on bone and bone graft healing due to the effects of osteoblast and osteoclast regulation and stimulation of angiogenesis. METHODS: Rat zygomatic arch critical-size bone defect model (5 mm) was used as the experimental model. Thirty-two Sprague-Dawley rats (64 zygomatic arches) were divided into 4 groups (16 zygomatic arches in each). In groups 1 and 2, defects were reconstructed with the bone grafts harvested from the other side, and the right arc was named as group 1, and the left was group 2. At group 1, 200 µM/300 µL dosage of DFO was injected at the zygomatic arch region starting at the seventh day preoperatively and lasting until the 45th day postoperatively. Group 2 animals were defined as the control group of group 1, and 0.9% NaCl injection was applied. In groups 3 and 4, there was no repair after the formation of defects, and the right arc region was treated with DFO, and left was treated with 0.9% NaCl for postoperative 45 days, respectively. Radiological (computed tomography), histological (hematoxylin-eosin), and biomechanical (3-point bending test) tests were used for the evaluation. RESULTS: In radiological evaluation, there was a statistically significant decrease (P < 0.05) in bone defect size in group 3 animals at the 4th, 8th, and 12th weeks, and bone graft volume showed a statistical difference at all weeks (P < 0.05). In histological evaluation, it was observed that there was an increase in osteoblast number and vascularity rates (P < 0.05) in the DFO-treated groups at all weeks. Biomechanical evaluation of the subjects showed increase in bone strength in group 1 animals at 12 weeks. CONCLUSIONS: In this study, it was shown that DFO treatment increased bone graft incorporation and healing in critical-size bone defects. In this aspect, we suggest that DFO can be used to increase graft incorporation in risky areas and reduce the defect size in patients who are not suitable for vascularized bone graft transfer.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Trasplante Óseo , Deferoxamina/farmacología , Sideróforos/farmacología , Cicatrización de Heridas/efectos de los fármacos , Cigoma/cirugía , Animales , Deferoxamina/administración & dosificación , Masculino , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Sideróforos/administración & dosificación , Trasplante Autólogo , Cigoma/efectos de los fármacos , Cigoma/fisiología
8.
Stroke ; 46(9): 2576-83, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26304864

RESUMEN

BACKGROUND AND PURPOSE: Iron chelation therapy is emerging as a novel neuroprotective strategy. The mechanisms of neuroprotection are diverse and include both neuronal and vascular pathways. We sought to examine the effect of iron chelation on cerebrovascular function in healthy aging and to explore whether hypoxia-inducible transcription factor-1 activation may be temporally correlated with vascular changes. METHODS: We assessed cerebrovascular function (autoregulation, vasoreactivity, and neurovascular coupling) and serum concentrations of vascular endothelial growth factor and erythropoietin, as representative measures of hypoxia-inducible transcription factor-1 activation, during 6 hours of deferoxamine infusion in 24 young and 24 older healthy volunteers in a randomized, blinded, placebo-controlled cross-over study design. Cerebrovascular function was assessed using the transcranial Doppler ultrasound. Vascular endothelial growth factor and erythropoietin serum protein assays were conducted using the Meso Scale Discovery platform. RESULTS: Deferoxamine elicited a strong age- and time-dependent increase in the plasma concentrations of erythropoietin and vascular endothelial growth factor, which persisted ≤3 hours post infusion (age effect P=0.04; treatment×time P<0.01). Deferoxamine infusion also resulted in a significant time- and age-dependent improvement in cerebral vasoreactivity (treatment×time P<0.01; age P<0.01) and cerebral autoregulation (gain: age×time×treatment P=0.04). CONCLUSIONS: Deferoxamine infusion improved cerebrovascular function, particularly in older individuals. The temporal association between improved cerebrovascular function and increased serum vascular endothelial growth factor and erythropoietin concentrations is supportive of shared hypoxia-inducible transcription factor-1-regulated pathways. Therefore, pharmacological activation of hypoxia-inducible transcription factor-1 to enhance cerebrovascular function may be a promising neuroprotective strategy in acute and chronic ischemic syndromes, especially in elderly patients. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT013655104.


Asunto(s)
Circulación Cerebrovascular/efectos de los fármacos , Terapia por Quelación/métodos , Deferoxamina/farmacología , Eritropoyetina/sangre , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Sideróforos/farmacología , Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/efectos de los fármacos , Estudios Cruzados , Deferoxamina/administración & dosificación , Femenino , Hemodinámica/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Sideróforos/administración & dosificación , Transducción de Señal , Resultado del Tratamiento , Ultrasonografía Doppler Transcraneal , Factores de Crecimiento Endotelial Vascular/sangre , Adulto Joven
9.
Mol Pharm ; 12(9): 3146-57, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26181293

RESUMEN

Excessive extracellular matrix formation in organs and tissues arises from an imbalance between the synthesis and degradation of matrix proteins, especially collagen. This condition interferes with proper wound healing and regeneration, and to date, no specific treatment is available. In the present study, we propose a targeted drug delivery system consisting of cell-specific immunoliposomes (ILs) loaded with deferoxamine (DFO) as an antifibrotic drug. ILs were functionalized with polyethylene glycol (PEG) to improve the steric stability and prolong their half-life. In addition, a single-chain Fv (scFv) antibody fragment that specifically targets fibroblast activation protein (FAP) was incorporated. An in vitro fibrosis model was employed to test this construct. This model consisted of highly activated pro-fibrotic fibroblasts with 2- to 6-fold induction of selected fibrosis markers: cell/matrix deposited collagen I, total soluble collagen, and α smooth muscle actin. The activation was accompanied by a significant and cell-specific elevation of FAP expression and activity, thereby confirming that FAP is an adequate target for antifibrotic drug delivery. Purified anti-FAP scFv was shown to bind specifically to these cells without influencing the FAP enzymatic activity. DFO was demonstrated to have a dose-dependent antifibrotic activity as quantified by collagen deposition. Specific binding and intracellular uptake of DiI-labeled ILs into the activated fibroblasts were shown by flow cytometry and microscopy. Finally, DFO-loaded ILs targeted to FAP caused a significant reduction in the collagen deposition, whereas no effect was observed using liposomes that lacked the targeting antibody fragment. These results suggest that the FAP-specific scFv-conjugated liposomes have considerable potential for cell-specific targeting applicable as a therapy for excessive collagen deposition during fibrosis. In general, through liposome encapsulation, bioactive molecules, such as DFO, that have broad effects and poor cell penetration can be converted into cell-specific composites for targeted drug delivery.


Asunto(s)
Deferoxamina/administración & dosificación , Fibroblastos/efectos de los fármacos , Fibrosarcoma/tratamiento farmacológico , Gelatinasas/antagonistas & inhibidores , Liposomas/química , Pulmón/efectos de los fármacos , Proteínas de la Membrana/antagonistas & inhibidores , Anticuerpos de Cadena Única/administración & dosificación , Células Cultivadas , Sistemas de Liberación de Medicamentos , Endopeptidasas , Fibroblastos/inmunología , Fibroblastos/patología , Fibrosarcoma/inmunología , Fibrosarcoma/patología , Gelatinasas/inmunología , Semivida , Humanos , Técnicas para Inmunoenzimas , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/inmunología , Liposomas/inmunología , Pulmón/inmunología , Pulmón/patología , Proteínas de la Membrana/inmunología , Polietilenglicoles/química , Serina Endopeptidasas/inmunología , Sideróforos/administración & dosificación , Anticuerpos de Cadena Única/inmunología
10.
Acta Haematol ; 133(2): 226-36, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25376266

RESUMEN

OBJECTIVE: To study the efficacy of combined treatment with oral and subcutaneous iron chelators. MATERIAL AND METHODS: 50-100 mg/kg/day of oral deferiprone (DFP) combined with 40 mg/kg/dose s.c. desferrioxamine (DFO) twice weekly were given to transfusion-dependent ß-thalassemia children. RESULTS: Enrolled patients (9 with ß-thalassemia major and 33 with ß-thalassemia hemoglobin E), ranging from 3 to 18 years in age, were divided into 3 groups; group 1 ferritin ≥1,000-2,500 ng/ml (n = 10), group 2 ferritin >2,500-4,000 ng/ml (n = 23) and group 3 ferritin >4,000 ng/ml (n = 9). Of the 42 patients, 28 reached the 36-month follow-up. Ten patients whose ferritin declined <15% while receiving 100 mg/kg/day of DFP were considered nonresponders. The median age and previous transfusion duration before enrollment were significantly higher in nonresponders than responders (p = 0.04 and 0.003, respectively). The responders exhibited a significant fall in median ferritin levels from 2,954.6 to 936.6 ng/ml (p < 0.001). Time to a significant decrease in serum ferritin among responders was 6 months. In 13 patients, 16 episodes of adverse events occurred: hemophagocytosis with cytopenia (n = 1), neutropenia (n = 2), thrombocytopenia (n = 2), elevated alanine aminotransferase (n = 5), elevated serum creatinine (n = 1), proteinuria (n = 1) and gastrointestinal discomfort (n = 4). CONCLUSION: Combination therapy with daily oral DFP and subcutaneous DFO twice weekly is a safe and effective alternative to chelation monotherapy in ß-thalassemia children.


Asunto(s)
Deferoxamina/administración & dosificación , Piridonas/administración & dosificación , Sideróforos/administración & dosificación , Talasemia beta/tratamiento farmacológico , Administración Oral , Adolescente , Alanina Transaminasa/sangre , Niño , Preescolar , Creatinina/sangre , Deferiprona , Deferoxamina/efectos adversos , Quimioterapia Combinada/métodos , Femenino , Ferritinas/sangre , Hemoglobina E/metabolismo , Humanos , Infusiones Subcutáneas , Masculino , Neutropenia/sangre , Neutropenia/inducido químicamente , Piridonas/efectos adversos , Sideróforos/efectos adversos , Trombocitopenia/sangre , Trombocitopenia/inducido químicamente , Talasemia beta/sangre
11.
Transfus Med ; 25(1): 8-12, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25801075

RESUMEN

BACKGROUND: The problem of iron-overload observed in thalassemia patients can be overcome using chelating agents such as deferiprone (Ferroprox(®) ), deferasirox (Exjade(®) ) and deferoxamine (Desferal(®) ). Although these drugs can be used as monotherapy, combined therapy, especially deferiprone with deferoxamine, has led to promising outcomes in various studies. METHODS AND MATERIALS: In this quasi-experimental study, serum ferritin levels were evaluated in 32 ß-thalassemia major patients with severe iron overload before and after receiving combined deferasirox (30-40 mg kg(-1) day(-1) ) and deferoxamine (40-50 mg kg(-1) day(-1) ) 2 days a week. This study was conducted from September 2012 to September 2013 in Southern Iran. RESULTS: The mean of serum ferritin levels significantly reduced from 4031 ± 1955 to 2416 ± 1653 ng mL(-1) after 12 months of therapy (P < 0·001). Echocardiograph findings showed significant improvement 1year after end of the study (P < 0·001). No drug toxicity was observed by monitoring serum creatinine, liver enzymes and blood urea nitrogen (BUN) during the study period. We observed no correlation between mean serum ferritin change and age (P = 0·87). In addition, the mean serum ferritin change did not differ between male and female thalassemia patients (P = 0·454). No difference in mean serum ferritin change was observed between patients who had undergone splenectomy compared to those who had not done so (P = 0·307). CONCLUSION: The study suggests that combination chelating therapy with deferasirox and deferoxamine can effectively reduce iron burden in ß-thalassemia major patients with heavy iron overload without any significant complications.


Asunto(s)
Benzoatos/administración & dosificación , Deferoxamina/administración & dosificación , Sideróforos/administración & dosificación , Triazoles/administración & dosificación , Talasemia beta/tratamiento farmacológico , Adolescente , Adulto , Niño , Deferasirox , Países en Desarrollo , Quimioterapia Combinada , Femenino , Ferritinas/sangre , Humanos , Irán , Líbano , Masculino , Talasemia beta/sangre
12.
Ann Plast Surg ; 74(2): 252-5, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25590247

RESUMEN

BACKGROUND: Prefabricated flaps are an ideal alternative to repair massive and complex tissue defects. Nevertheless, the risk of necrosis due to unpredictable blood supplies is a major obstacle to the application of prefabricated flaps. The survival of a prefabricated flap depends on the neovascularization between the vascular carrier and the donor tissue. Here, we proposed that the iron chelator, desferrioxamine (DFX), owned therapeutic effects that promoted the neovascularization of prefabricated flaps. METHODS: An abdominal prefabricated flap model was created in rats via a 2-stage operation. The rats were allocated into 4 groups as follows: 2 groups of rats received DFX treatments during the first or the second stage of the operation, respectively; 1 group of rats received a delay procedure 1 week before the second operation; and the final group was used as a blank control. Flap survival rates and capillary densities were evaluated between groups. The influence of DFX on the dermal fibroblasts was also studied in vitro. RESULTS: Desferrioxamine treatment during the first stage of the operation greatly increased flap survival rate compared to the blank control. The results were similar to those produced by the delay treatment. The vessel count results were consistent with the flap survival rate findings. In vitro, DFX treatment up-regulated the expression levels of several angiogenic factors in the dermal fibroblasts. Nevertheless, DFX treatment during the second stage of the operation was therapeutically detrimental. CONCLUSIONS: The application of DFX around the time of vascular carrier implantation greatly promoted neovascularization of prefabricated flaps, but was therapeutically detrimental after the flaps had been elevated.


Asunto(s)
Deferoxamina/farmacología , Supervivencia de Injerto/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Sideróforos/farmacología , Colgajos Quirúrgicos/irrigación sanguínea , Animales , Capilares/anatomía & histología , Capilares/efectos de los fármacos , Deferoxamina/administración & dosificación , Esquema de Medicación , Inyecciones Intraperitoneales , Masculino , Distribución Aleatoria , Ratas , Ratas Wistar , Sideróforos/administración & dosificación
13.
BMC Ophthalmol ; 14: 88, 2014 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-24989140

RESUMEN

BACKGROUND: To describe the spectral domain optical coherence tomography (SD-OCT) findings of a patient who developed pigmentary retinopathy following high-dose deferoxamine administration. CASE PRESENTATION: A 34-year-old man with thalassemia major complained of nyctalopia and decreased vision following high-dose intravenous deferoxamine to treat systemic iron overload. Fundus examination revealed multiple discrete hypo-pigmented lesions at the posterior pole and mid-peripheral retina. Recovery was partial following cessation of desferrioxamine six weeks later. A follow-up SD-OCT showed multiple accumulated hyper-reflective deposits primarily in the choroid, retina pigment epithelium (RPE), and inner segment and outer segment (IS/OS) junction. CONCLUSION: Deferoxamine retinopathy primarily targets the RPE-Bruch membrane-photoreceptor complex, extending from the peri-fovea to the peripheral retina with foveola sparing. An SD-OCT examination can serve as a simple, noninvasive tool for early detection and long-term follow-up.


Asunto(s)
Deferoxamina/efectos adversos , Enfermedades de la Retina/diagnóstico , Epitelio Pigmentado de la Retina/patología , Tomografía de Coherencia Óptica/métodos , Adulto , Deferoxamina/administración & dosificación , Relación Dosis-Respuesta a Droga , Angiografía con Fluoresceína , Fondo de Ojo , Humanos , Inyecciones Intravenosas , Masculino , Enfermedades de la Retina/inducido químicamente , Epitelio Pigmentado de la Retina/efectos de los fármacos , Sideróforos/administración & dosificación , Sideróforos/efectos adversos , Talasemia/tratamiento farmacológico , Agudeza Visual
14.
J Med Assoc Thai ; 97 Suppl 8: S217-22, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25518318

RESUMEN

Thalassemia major is an inherited form of chronic hemolytic anemia that results in iron overload due to regular blood transfusions. Deferoxamine is used as chelating agentfor treatment ofpatients with chronic iron overload worldwide. Anaphylactic reaction to deferoxamine is rare, and the mechanism ofdeferoxamine-induced anaphylaxis is not well understood. Only afewpediatric cases ofsuccessful desensitization for deferoxamine hypersensitivity have been described, and a different protocol has been used in each report. We report a case ofanaphylaxis to deferoxamine in a thirteen-years-old Thai boy with Hemoglobin E/ß-thalassemia disease who underwent successful desensitization. He had been receiving blood transfusions since the age often months. At age eleven, the patient began treatment with deferoxamine. Treatment was interrupted after the occurrence ofanaphylaxis, with urticaria, wheezing and gastrointestinal symptoms. A skin prick test was positive, indicating a type 1 hypersensitivity reaction. Deferoxamine desensitization was attempted with various differentprotocols. Finally, the patient could tolerate deferoxamine therapy at the dose previously administered. We proposed this modified subcutaneous desensitization protocolforpediatric cases that develop allergic reactions to deferoxamine.


Asunto(s)
Anafilaxia/prevención & control , Deferoxamina/administración & dosificación , Desensibilización Inmunológica/métodos , Sobrecarga de Hierro/tratamiento farmacológico , Sideróforos/administración & dosificación , Talasemia beta/terapia , Adolescente , Anafilaxia/inducido químicamente , Transfusión Sanguínea , Deferoxamina/efectos adversos , Hipersensibilidad a las Drogas/etiología , Hipersensibilidad a las Drogas/prevención & control , Humanos , Hipersensibilidad Inmediata/inducido químicamente , Hipersensibilidad Inmediata/prevención & control , Inyecciones Subcutáneas , Masculino , Sideróforos/efectos adversos , Pruebas Cutáneas
15.
Lakartidningen ; 111(38): 1576-7, 2014.
Artículo en Sueco | MEDLINE | ID: mdl-25606656

RESUMEN

A 20-year-old woman was found semiconscious on the floor in a pool of black diarrhea with an empty 100 jar of ferrous sulphate beside her (100 mg Fe2+/tablet), 160 mg/kg. She was brought to the hospital an estimated 4 hours after ingestion and presented with irritability and a fluctuating CNS depression. Her blood pressure was 190/85 mmHg and pulse 130 bpm. An arterial blood gas analysis showed pH 7.17, pCO2 5.4 kPa, pO2 16.7 kPa and BE ­14 mmol/l. Deferoxamine was started immediately with a dose of 15 mg/kg/h intravenously. The patient was intubated in the ICU and whole bowel irrigation was performed. Due to technical problems with the venous blood sampling, a correct measurement of the serum iron concentration (s-Fe2) was not at hand until 15 hours post ingestion and showed 131 µmol/l. At that point her condition deteriorated with circulatory instability, hepatic failure, coagulopathy and renal insufficiency. Despite full treatment including continuous renal replacement therapy she died after 4 Days.


Asunto(s)
Sobredosis de Droga/complicaciones , Compuestos Ferrosos/envenenamiento , Sobrecarga de Hierro/complicaciones , Adulto , Deferoxamina/administración & dosificación , Deferoxamina/uso terapéutico , Sobredosis de Droga/tratamiento farmacológico , Sobredosis de Droga/terapia , Resultado Fatal , Femenino , Humanos , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/terapia , Sideróforos/administración & dosificación , Sideróforos/uso terapéutico , Suicidio
16.
Bioconjug Chem ; 24(3): 473-86, 2013 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-23350642

RESUMEN

Siderophores are multidentate iron(III) chelators used by bacteria for iron assimilation. Sideromycins, also called siderophore-antibiotic conjugates, are a unique subset of siderophores that enter bacterial cells via siderophore uptake pathways and deliver the toxic antibiotic in a "Trojan horse" fashion. Sideromycins represent a novel antibiotic delivery technology with untapped potential for developing sophisticated microbe-selective antibacterial agents that limit the emergence of bacterial resistance. The chemical synthesis of a series of mono-, bis-, and trihydroxamate sideromycins are described here along with their biological evaluation in antibacterial susceptibility assays. The linear hydroxamate siderophores used for the sideromycins in this study were derived from the ferrioxamine family and inspired by the naturally occurring salmycin sideromycins. The antibacterial agents used were a ß-lactam carbacepholosporin, Lorabid, and a fluoroquinolone, ciprofloxacin, chosen for the different locations of their biological targets, the periplasm (extracellular) and the cytoplasm (intracellular). The linear hydroxamate-based sideromycins were selectively toxic toward Gram-positive bacteria, especially Staphylococcus aureus SG511 (MIC = 1.0 µM for the trihydroxamate-fluoroquinolone sideromycin). Siderophore-sideromycin competition assays demonstrated that only the fluoroquinolone sideromycins required membrane transport to reach their cytoplasmic biological target and that a trihydroxamate siderophore backbone was required for protein-mediated active transport of the sideromycins into S. aureus cells via siderophore uptake pathways. This work represents a comprehensive study of linear hydroxamate sideromycins and teaches how to build effective hydroxamate-based sideromycins as Gram-positive selective antibiotic agents.


Asunto(s)
Antibacterianos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Compuestos Ferrosos/administración & dosificación , Fluoroquinolonas/administración & dosificación , Ácidos Hidroxámicos/administración & dosificación , Péptidos/administración & dosificación , Sideróforos/administración & dosificación , Staphylococcus aureus/efectos de los fármacos , Antibacterianos/química , Antibacterianos/metabolismo , Compuestos Ferrosos/química , Compuestos Ferrosos/metabolismo , Fluoroquinolonas/química , Fluoroquinolonas/metabolismo , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/metabolismo , Pruebas de Sensibilidad Microbiana , Péptidos/química , Péptidos/metabolismo , Sideróforos/química , Sideróforos/metabolismo , Staphylococcus aureus/metabolismo
17.
Cochrane Database Syst Rev ; (8): CD004450, 2013 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-23963793

RESUMEN

BACKGROUND: Thalassaemia major is a genetic disease characterised by a reduced ability to produce haemoglobin. Management of the resulting anaemia is through red blood cell transfusions.Repeated transfusions result in an excessive accumulation of iron in the body (iron overload), removal of which is achieved through iron chelation therapy. Desferrioxamine mesylate (desferrioxamine) is one of the most widely used iron chelators. Substantial data have shown the beneficial effects of desferrioxamine, although adherence to desferrioxamine therapy is a challenge. Alternative oral iron chelators, deferiprone and deferasirox, are now commonly used. Important questions exist about whether desferrioxamine, as monotherapy or in combination with an oral iron chelator, is the best treatment for iron chelation therapy. OBJECTIVES: To determine the effectiveness (dose and method of administration) of desferrioxamine in people with transfusion-dependent thalassaemia.To summarise data from trials on the clinical efficacy and safety of desferrioxamine for thalassaemia and to compare these with deferiprone and deferasirox. SEARCH METHODS: We searched the Cochrane Cystic Fibrosis and Genetic Disorders Group's Haemoglobinopathies Trials Register. We also searched MEDLINE, EMBASE, CENTRAL (The Cochrane Library), LILACS and other international medical databases, plus ongoing trials registers and the Transfusion Evidence Library (www.transfusionevidencelibrary.com). All searches were updated to 5 March 2013. SELECTION CRITERIA: Randomised controlled trials comparing desferrioxamine with placebo, with another iron chelator, or comparing two schedules or doses of desferrioxamine, in people with transfusion-dependent thalassaemia. DATA COLLECTION AND ANALYSIS: Six authors working independently were involved in trial quality assessment and data extraction. For one trial, investigators supplied additional data upon request. MAIN RESULTS: A total of 22 trials involving 2187 participants (range 11 to 586 people) were included. These trials included eight comparisons between desferrioxamine alone and deferiprone alone; five comparisons between desferrioxamine combined with deferiprone and deferiprone alone; eight comparisons between desferrioxamine alone and desferrioxamine combined with deferiprone; two comparisons of desferrioxamine with deferasirox; and two comparisons of different routes of desferrioxamine administration (bolus versus continuous infusion). Overall, few trials measured the same or long-term outcomes. Seven trials reported cardiac function or liver fibrosis as measures of end organ damage; none of these included a comparison with deferasirox.Five trials reported a total of seven deaths; three in patients who received desferrioxamine alone, two in patients who received desferrioxamine and deferiprone. A further death occurred in a patient who received deferiprone in another who received deferasirox alone. One trial reported five further deaths in patients who withdrew from randomised treatment (deferiprone with or without desferrioxamine) and switched to desferrioxamine alone.One trial planned five years of follow up but was stopped early due to the beneficial effects of a reduction in serum ferritin levels in those receiving combined desferrioxamine and deferiprone treatment compared with deferiprone alone. The results of this and three other trials suggest an advantage of combined therapy with desferrioxamine and deferiprone over monotherapy to reduce iron stores as measured by serum ferritin. There is, however, no evidence for the improved efficacy of combined desferrioxamine and deferiprone therapy against monotherapy from direct or indirect measures of liver iron.Earlier trials measuring the cardiac iron load indirectly by measurement of the magnetic resonance imaging T2* signal had suggested deferiprone may reduce cardiac iron more quickly than desferrioxamine. However, meta-analysis of two trials showed a significantly lower left ventricular ejection fraction in patients who received desferrioxamine alone compared with those who received combination therapy using desferrioxamine with deferiprone.Adverse events were recorded by 18 trials. These occurred with all treatments, but were significantly less likely with desferrioxamine than deferiprone in one trial, relative risk 0.45 (95% confidence interval 0.24 to 0.84) and significantly less likely with desferrioxamine alone than desferrioxamine combined with deferiprone in two other trials, relative risk 0.33 (95% confidence interval 0.13 to 0.84). In particular, four studies reported permanent treatment withdrawal due to adverse events from deferiprone; only one of these reported permanent withdrawals associated with desferrioxamine. Adverse events also occurred at a higher frequency in patients who received deferasirox than desferrioxamine in one trial. Eight trials reported local adverse reactions at the site of desferrioxamine infusion including pain and swelling. Adverse events associated with deferiprone included joint pain, gastrointestinal disturbance, increases in liver enzymes and neutropenia; adverse events associated with deferasirox comprised increases in liver enzymes and renal impairment. Regular monitoring of white cell counts has been recommended for deferiprone and monitoring of liver and renal function for deferasirox.In summary, desferrioxamine and the oral iron chelators deferiprone and deferasirox produce significant reductions in iron stores in transfusion-dependent, iron-overloaded people. There is no evidence from randomised clinical trials to suggest that any one of these has a greater reduction of clinically significant end organ damage, although in two trials, combination therapy with desferrioxamine and deferiprone showed a greater improvement in left ventricular ejection fraction than desferrioxamine used alone. AUTHORS' CONCLUSIONS: Desferrioxamine is the recommended first-line therapy for iron overload in people with thalassaemia major and deferiprone or deferasirox are indicated for treating iron overload when desferrioxamine is contraindicated or inadequate. Oral deferasirox has been licensed for use in children aged over six years who receive frequent blood transfusions and in children aged two to five years who receive infrequent blood transfusions. In the absence of randomised controlled trials with long-term follow up, there is no compelling evidence to change this conclusion.Worsening iron deposition in the myocardium in patients receiving desferrioxamine alone would suggest a change of therapy by intensification of desferrioxamine treatment or the use of desferrioxamine and deferiprone combination therapy.Adverse events are increased in patients treated with deferiprone compared with desferrioxamine and in patients treated with combined deferiprone and desferrioxamine compared with desferrioxamine alone. People treated with all chelators must be kept under close medical supervision and treatment with deferiprone or deferasirox requires regular monitoring of neutrophil counts or renal function respectively. There is an urgent need for adequately-powered, high-quality trials comparing the overall clinical efficacy and long-term outcomes of deferiprone, deferasirox and desferrioxamine.


Asunto(s)
Deferoxamina/administración & dosificación , Quelantes del Hierro/administración & dosificación , Sobrecarga de Hierro/tratamiento farmacológico , Sideróforos/administración & dosificación , Talasemia/terapia , Reacción a la Transfusión , Benzoatos/administración & dosificación , Terapia por Quelación , Deferasirox , Deferiprona , Humanos , Sobrecarga de Hierro/etiología , Piridonas/administración & dosificación , Ensayos Clínicos Controlados Aleatorios como Asunto , Triazoles/administración & dosificación
18.
Neurocrit Care ; 19(2): 257-66, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23943316

RESUMEN

BACKGROUND: Hemoglobin degradation products, in particular iron, have been implicated in secondary neuronal injury following intracerebral hemorrhage (ICH). The iron chelator Deferoxamine Mesylate (DFO) exerts diverse neuroprotective effects, reduces perihematoma edema (PHE) and neuronal damage, and improves functional recovery after experimental ICH. We hypothesize that treatment with DFO could minimize neuronal injury and improve outcome in ICH patients. As a prelude to test this hypothesis, we conducted a Phase I, open-label study to determine the tolerability, safety, and maximum tolerated dose (MTD) of DFO in patients with ICH. Intravenous infusions of DFO in doses up to 62 mg/kg/day (up to a maximum of 6000 mg/day) were well-tolerated and did not seem to increase serious adverse events (SAEs) or mortality. We have initiated a multi-center, double-blind, randomized, placebo-controlled, Phase II clinical trial (High Dose Deferoxamine [HI-DEF] in Intracerebral Hemorrhage) to determine if it is futile to move DFO forward to Phase III efficacy evaluation. METHODS: We will randomize 324 subjects with spontaneous ICH to either DFO at 62 mg/kg/day (up to a maximum daily dose of 6000 mg/day) or saline placebo, given by intravenous infusion for 5 consecutive days. Treatment will be initiated within 24 hours after ICH symptom onset. All subjects will be followed for 3 months and will receive standard of care therapy while participating in the study. At 3 months, the proportion of DFO-treated subjects with a good clinical outcome, assessed by modified Rankin Scale, will be compared to the placebo proportion in a futility analysis. CONCLUSIONS: The Hi-Def trial is expected to advance our understanding of the pathopgysiology of secondary neuronal injury in ICH and will provide a crucial "Go/No Go" signal as to whether a Phase III trial to investigate the efficacy of DFO is warranted.


Asunto(s)
Hemorragia Cerebral/tratamiento farmacológico , Cuidados Críticos/métodos , Deferoxamina/administración & dosificación , Proyectos de Investigación , Sideróforos/administración & dosificación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Hemorragia Cerebral/fisiopatología , Deferoxamina/efectos adversos , Método Doble Ciego , Humanos , Persona de Mediana Edad , Placebos , Sideróforos/efectos adversos , Adulto Joven
20.
Neurol Sci ; 33(2): 289-96, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21863269

RESUMEN

Deferoxamine (DFX), a potent iron-chelating agent, reduces brain edema and neuronal cell injury that develop due to the hemolysis cascade. Statins have neuroprotective effects via anti-inflammatory action and increment of cerebral blood flow after intracerebral hemorrhage (ICH). The purpose of this study was to identify the effects of combined DFX and statins treatment in an experimental ICH rat model. The treatments were: intraperitoneal (i.p.) injection of DFX (group I), combined treatment of i.p. DFX and oral statins (group II), statins only (group III) and treatment with vehicle (group IV). Induction of ICH was performed with injection of bacterial collagenase type IV into the left striatum. After removal of the brain, hematoma volume, water content and brain atrophy were measured. Immunohistochemistry in the perihematomal region was performed for identification of microglial infiltration, astrocyte expression and apoptotic cell presence. Statistical analysis was performed using the non-parametric Kruskal-Wallis test and significance was evaluated when the p value was less than 0.05. According to behavioral tests, significant differences among treatment groups were noted 4 weeks after ICH induction (p < 0.05). However, there were no significant differences among treatment groups in hematoma volume, brain water content or brain atrophy. In the perihematomal area, the activated microglial cells were reduced in the combined treatment group. Among the four groups, a significant difference in immunohistochemical staining was identified (p < 0.05). These results suggest that combined treatment with DFX and statins improves neurologic outcomes after ICH through reduction of microglial infiltration, apoptosis, inflammation and brain edema.


Asunto(s)
Hemorragia Cerebral/complicaciones , Deferoxamina/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/etiología , Sideróforos/administración & dosificación , Análisis de Varianza , Animales , Atrofia/tratamiento farmacológico , Atrofia/etiología , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Antígeno CD11b/metabolismo , Modelos Animales de Enfermedad , Extremidades/fisiopatología , Proteína Ácida Fibrilar de la Glía/metabolismo , Hematoma/tratamiento farmacológico , Hematoma/etiología , Masculino , Desempeño Psicomotor/efectos de los fármacos , Desempeño Psicomotor/fisiología , Ratas , Ratas Sprague-Dawley , Conducta Espacial/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA