Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
1.
Nanomedicine (Lond) ; 17(22): 1649-1662, 2022 09.
Article in English | MEDLINE | ID: mdl-36547231

ABSTRACT

Aim: To characterize the pharmacokinetics of deferoxamine-conjugated nanoparticles (DFO-NPs), a novel nanochelator for removing excess iron. Materials & methods: The pharmacokinetics of DFO-NPs were evaluated in Sprague-Dawley rats at three doses (3.3, 10 and 30 µmol/kg) after intravenous and subcutaneous administration. Results: DFO-NPs exhibited a biphasic concentration-time profile after intravenous administration with a short terminal half-life (2.0-3.2 h), dose-dependent clearance (0.111-0.179 l/h/kg), minimal tissue distribution and exclusive renal excretion with a possible saturable reabsorption mechanism. DFO-NPs after subcutaneous administration exhibited absorption-rate-limited kinetics with a prolonged half-life (5.7-10.1 h) and favorable bioavailability (47-107%). Conclusion: DFO-NPs exhibit nonlinear pharmacokinetics with increasing dose, and subcutaneous administration substantially improves drug exposure, thereby making it a clinically viable administration route for iron chelation.


Iron is an essential metal nutrient, but excess iron produces toxic effects that damage multiple organs including the heart, liver and pancreas. Deferoxamine (DFO) is a US FDA-approved drug for treating iron overload, but its use is limited by serious adverse effects and an inconvenient daily dose scheme. The recent development of a DFO-based nanomedicine (DFO-NP) has shown promise in treating iron overload in animals and was safer in animals. Before this new drug can be given to humans, how it is absorbed into the body, processed in the body and removed from the body when given in different amounts and dose routes must be determined. In this study, we tested the absorption, distribution and removal of DFO-NPs after intravenous and subcutaneous injection in rats. This study showed that DFO-NPs behave differently when changing the dose and that subcutaneous injection makes the drug stay in the body longer without ill effect, which means it could be given to patients this way.


Subject(s)
Deferoxamine , Iron Overload , Rats , Animals , Deferoxamine/pharmacokinetics , Deferoxamine/therapeutic use , Iron Chelating Agents/pharmacokinetics , Iron Chelating Agents/therapeutic use , Tissue Distribution , Rats, Sprague-Dawley , Iron Overload/drug therapy
2.
Ann Hematol ; 101(3): 533-539, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34981144

ABSTRACT

Patients with sickle cell disease (SCD) who undergo repeated blood transfusions often develop iron overload. Deferiprone (Ferriprox®) is an oral iron chelator indicated for the treatment of transfusional iron overload due to thalassemia syndromes and has been recently approved as a treatment for iron overload in adult and pediatric patients with SCD and other anemias. The present study aims to characterize the pharmacokinetic (PK) profile of deferiprone (DFP) in adult subjects with SCD. In this phase I, open-label study, subjects with SCD were administered a single 1500 mg dose of DFP. Blood and urine samples were collected for PK assessments of DFP and its main metabolite, deferiprone 3-O-glucuronide (DFP-G). Eight subjects were enrolled and completed the study. Following drug administration, serum levels of DFP and DFP-G rose to maximum concentrations at 1.0 and 2.8 h post-dose, respectively. The half-lives of DFP and DFP-G were 1.5 and 1.6 h, respectively. The majority of administered drug was metabolized and excreted as DFP-G, with less than 4% excreted unchanged in urine up to 10 h post-dose. Subjects received a safety assessment 7 (± 3) days post-dose. Two subjects reported mild adverse events unrelated to the study drug, and no other safety concerns were reported. The PK profile of DFP in SCD subjects is consistent with previous reports in healthy adult volunteers, suggesting no special dosing adjustments are indicated for this population. These findings provide valuable insight for treating iron overload in patients with SCD, who have limited chelation therapy treatment options (trial registration number: NCT01835496, date of registration: April 19, 2013).


Subject(s)
Anemia, Sickle Cell/complications , Deferiprone/pharmacokinetics , Iron Chelating Agents/pharmacokinetics , Iron Overload/drug therapy , Adult , Anemia, Sickle Cell/therapy , Blood Transfusion , Chelation Therapy/adverse effects , Deferiprone/adverse effects , Deferiprone/therapeutic use , Female , Humans , Iron Chelating Agents/adverse effects , Iron Chelating Agents/therapeutic use , Iron Overload/etiology , Male , Young Adult
3.
Sci Rep ; 11(1): 12581, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34131221

ABSTRACT

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


Subject(s)
Anemia/drug therapy , Deferasirox/administration & dosage , Iron Overload/drug therapy , Thalassemia/drug therapy , Adult , Anemia/blood , Anemia/epidemiology , Anemia/pathology , Chelation Therapy/trends , Deferasirox/pharmacokinetics , Female , Ferritins/blood , Humans , Iron/blood , Iron/metabolism , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/pharmacokinetics , Iron Overload/blood , Iron Overload/epidemiology , Iron Overload/pathology , Male , Middle Aged , Retrospective Studies , Thalassemia/blood , Thalassemia/epidemiology , Thalassemia/pathology
4.
Eur J Drug Metab Pharmacokinet ; 45(6): 761-770, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32930952

ABSTRACT

BACKGROUND AND OBJECTIVE: Deferasirox is an oral iron chelator used to reduce iron levels in iron-overloaded patients with transfusion-dependent anemia or non-transfusion-dependent thalassemia. This study investigated the effects of genetic polymorphisms on the pharmacokinetics of deferasirox in healthy Chinese subjects and constructed a pharmacokinetic prediction model based on physiologic factors and genetic polymorphism data. METHODS: Twenty-eight subjects were enrolled in a randomized, open-label, two-period crossover study, and they received a single dose of one of two formulations of deferasirox (20 mg/kg) with a 7-day washout interval between the two periods. The plasma defersirox concentration was determined using a validated liquid chromatography-tandem mass spectrometry method, and pharmacokinetic parameters were calculated using the noncompartmental method. The polymorphisms of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1), UGT1A3, multidrug resistance protein 2 (MRP2), cytochrome P450 1A1 (CYP1A1), and breast cancer resistance protein 1 (BCRP1) were genotyped using Sanger sequencing. A back-propagation artificial neural network (BP-ANN) model was used to predict the pharmacokinetics. RESULTS: The UGT1A1 rs887829 C > T single-nucleotide polymorphism (SNP) significantly influenced the area under the plasma concentration-time curve and the terminal half-life. Neither the MRP2 rs2273697 G > A SNP nor BCRP1 rs2231142 G > T SNP altered the absorption, disposition, and excretion of the drug. The BP-ANN model had a high goodness-of-fit index and good coherence between the predicted and measured concentrations (R2 = 0.921). CONCLUSION: Metabolic enzyme-related genetic polymorphisms were more strongly associated with the pharmacokinetics of deferasirox than membrane transporter-related genetic polymorphisms in the Chinese population. TRIAL REGISTRATION: www.Chinadrugtrials.org.cn CTR20191164.


Subject(s)
Deferasirox/pharmacokinetics , Iron Chelating Agents/pharmacokinetics , Neural Networks, Computer , Adolescent , Adult , Area Under Curve , Asian People , Cross-Over Studies , Cytochrome P-450 CYP1A1/genetics , Drug Compounding , Female , Glucuronosyltransferase/genetics , Half-Life , Healthy Volunteers , Humans , Male , Middle Aged , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Polymorphism, Genetic/genetics , Polymorphism, Single Nucleotide , Predictive Value of Tests , Young Adult
5.
Biochim Biophys Acta Gen Subj ; 1864(10): 129670, 2020 10.
Article in English | MEDLINE | ID: mdl-32565293

ABSTRACT

BACKGROUND: Microorganisms produce siderophores in order to scavenge iron from the environment and this study focuses on the characterization of the two siderophores secreted by Bacillus megaterium. The general biological properties and pharmacokinetics following oral application of these compounds are reported. METHODS: Under optimized culture conditions, the siderophores were harvested, purified by chromatography and identified using LC-MS and NMR. Two dihydroxamate siderophores were isolated, schizokinen (MW = 420) and schizokinen imide (MW = 402). RESULTS: Both compounds demonstrate strong antioxidant activity and were found to be relatively nontoxic to both human hepatocellular carcinoma (Huh7) and peripheral blood mononuclear cells. The siderophores possess a strong affinity for iron(III) and decrease the levels of the labile iron pool (LIP) in iron-loaded cells in a concentration-dependent manner. Schizokinen, was detected as both the free siderophore and the iron complex in the plasma and urine of rats after oral gavage. CONCLUSIONS: However, the bioavailability was low and thus schizokinen, like deferoxamine, has no potential as an orally active iron chelator for the treatment of systemic iron overload. GENERAL SIGNIFICANCE: By virtue of the high affinity of schizokinen for tribasic metals, this siderophore does have considerable potential for the chelation of gallium(III) and the development of clinical diagnostic reagents.


Subject(s)
Antioxidants/chemistry , Antioxidants/pharmacology , Bacillus megaterium/chemistry , Siderophores/chemistry , Siderophores/pharmacology , Animals , Antioxidants/pharmacokinetics , Cell Line, Tumor , Cells, Cultured , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/pharmacology , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacokinetics , Iron Chelating Agents/pharmacology , Male , Rats, Sprague-Dawley , Siderophores/pharmacokinetics
6.
Basic Res Cardiol ; 115(3): 24, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32140789

ABSTRACT

Intramyocardial hemorrhage is an independent predictor of adverse outcomes in ST-segment elevation myocardial infarction (STEMI). Iron deposition resulting from ischemia-reperfusion injury (I/R) is pro-inflammatory and has been associated with adverse remodeling. The role of iron chelation in hemorrhagic acute myocardial infarction (AMI) has never been explored. The purpose of this study was to investigate the cardioprotection offered by the iron-chelating agent deferiprone (DFP) in a porcine AMI model by evaluating hemorrhage neutralization and subsequent cardiac remodeling. Two groups of animals underwent a reperfused AMI procedure: control and DFP treated (N = 7 each). A comprehensive MRI examination was performed in healthy state and up to week 4 post-AMI, followed by histological assessment. Infarct size was not significantly different between the two groups; however, the DFP group demonstrated earlier resolution of hemorrhage (by T2* imaging) and edema (by T2 imaging). Additionally, ventricular enlargement and myocardial hypertrophy (wall thickness and mass) were significantly smaller with DFP, suggesting reduced adverse remodeling, compared to control. The histologic results were consistent with the MRI findings. To date, there is no effective targeted therapy for reperfusion hemorrhage. Our proof-of-concept study is the first to identify hemorrhage-derived iron as a therapeutic target in I/R and exploit the cardioprotective properties of an iron-chelating drug candidate in the setting of AMI. Iron chelation could potentially serve as an adjunctive therapy in hemorrhagic AMI.


Subject(s)
Cardiotonic Agents/pharmacology , Deferiprone/therapeutic use , Hemorrhage/drug therapy , Hemorrhage/etiology , Iron Chelating Agents/therapeutic use , Myocardial Infarction/complications , Myocardium/pathology , Ventricular Remodeling/drug effects , Animals , Cardiotonic Agents/pharmacokinetics , Cardiotonic Agents/therapeutic use , Deferiprone/pharmacokinetics , Deferiprone/pharmacology , Disease Models, Animal , Female , Hemorrhage/pathology , Iron Chelating Agents/pharmacokinetics , Iron Chelating Agents/pharmacology , Myocardial Infarction/pathology , Swine
7.
J Inorg Biochem ; 193: 152-165, 2019 04.
Article in English | MEDLINE | ID: mdl-30769225

ABSTRACT

This work presents the simple and low cost synthesis of a new tripodal ligand, in which three units of kojic acid are coupled to a tris(2-aminoethyl)amine (tren) backbone molecule. The protonation equilibria, together with the complex formation equilibria of this ligand with Fe3+, Al3+, Cu2+ and Zn2+ ions were studied. The complementary use of potentiometric, spectrophotometric and NMR techniques, and of Density Functional Theory (DFT) calculations, has allowed a thorough characterization of the different species involved in equilibrium. The stability of the formed complexes with Fe3+ and Al3+ are high enough to consider the new ligand for further studies for its clinical applications as a chelating agent. Biodistribution studies were carried out to assess the capacity the ligand for mobilization of gallium in 67Ga-citrate injected mice. These studies demonstrated that this ligand efficiently chelates the radiometal in our animal model, which suggests that it can be a promising candidate as sequestering agent of iron and other hard trivalent metal ions. Furthermore, the good zinc complexation capacity appears as a stimulating result taking into a potential use of this new ligand in analytical chemistry as well as in agricultural and environmental applications.


Subject(s)
Iron Chelating Agents/pharmacology , Pyrans/pharmacology , Pyrones/pharmacology , Aluminum/chemistry , Animals , Copper/chemistry , Density Functional Theory , Female , Gallium Radioisotopes/chemistry , Iron/chemistry , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/pharmacokinetics , Mice , Models, Chemical , Pyrans/chemical synthesis , Pyrans/pharmacokinetics , Pyrones/chemical synthesis , Pyrones/pharmacokinetics , Tissue Distribution , Zinc/chemistry
8.
Invest New Drugs ; 37(4): 684-692, 2019 08.
Article in English | MEDLINE | ID: mdl-30460505

ABSTRACT

Introduction VLX600 is a novel iron chelator designed to interfere with intracellular iron metabolism, leading to inhibition of mitochondrial respiration and bioenergetic catastrophe and resultant tumor cell death. Methods We conducted a multicenter, phase 1, dose escalation study to determine the safety and adverse event profile and the maximum tolerated dose and recommended phase 2 dose of VLX600. Other endpoints included pharmacokinetics, and preliminary evidence of anti-cancer efficacy as assessed according to RECIST 1.1 criteria. VLX600 was administered intravenously on days 1, 8, and 15 of each 28-day treatment cycle. Results Nineteen patients were enrolled, and seventeen received at least one dose of VLX600. Dose increments were reduced to 50% after dose level 3 (40 mg) due to the occurrence of a grade 3 pulmonary embolism. The study was then closed early due to slow recruitment. No maximum tolerated dose (MTD) nor RP2D had been identified at the time of study closure. Overall, the drug was well tolerated and no DLTs were observed. Fourteen patients experienced drug-related adverse events of any grade. The most frequently reported drug-related AEs were fatigue, nausea, constipation, vomiting, increased alkaline phosphatase, anemia, and decreased appetite. No formal efficacy or survival analyses were performed. No objective responses were observed, though six patients (32%) had stable disease as best response. Conclusion VLX600 was reasonably well tolerated and, together with preclinical data, there is support for further efforts to explore its activity as single agent and in combination with drugs or radiation.


Subject(s)
Antineoplastic Agents/administration & dosage , Hydrazones/administration & dosage , Iron Chelating Agents/administration & dosage , Neoplasms/drug therapy , Triazoles/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Drug Resistance, Neoplasm , Female , Humans , Hydrazones/adverse effects , Hydrazones/blood , Hydrazones/pharmacokinetics , Iron Chelating Agents/adverse effects , Iron Chelating Agents/pharmacokinetics , Male , Middle Aged , Neoplasms/metabolism , Triazoles/adverse effects , Triazoles/blood , Triazoles/pharmacokinetics
10.
Int J Pharm ; 538(1-2): 79-86, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29341909

ABSTRACT

Deferoxamine (DFO) to treat iron overload (IO) has been limited by toxicity issues and short circulation times and it would be desirable to prolong circulation to improve non-transferrin bound iron (NTBI) chelation. In addition, DFO is currently unable to efficiently target the large pool of iron in the liver and spleen. Nanogel-Deferoxamine conjugates (NG-DFO) can prove useful as a model to investigate the pharmacokinetic (PK) properties and biodistribution (BD) behavior of iron-chelating macromolecules and their overall effect on serum ferritin levels. NG-DFO reduced the cytotoxicity of DFO and significantly reduced cellular ferritin levels in IO macrophages in vitro. PK/BD studies in normal rats revealed that NG-DFO displayed prolonged circulation and preferential accumulation into the liver and spleen. IO mice treated with NG1-DFO presented significantly lower levels of serum ferritin compared to DFO. Total renal and fecal elimination data point to the need to balance prolonged circulation with controlled degradation to accelerate clearance of iron-chelating macromolecules.


Subject(s)
Deferoxamine/administration & dosage , Iron Chelating Agents/administration & dosage , Iron Overload/drug therapy , Models, Biological , Animals , Deferoxamine/pharmacokinetics , Deferoxamine/pharmacology , Disease Models, Animal , Female , Ferritins/blood , Human Umbilical Vein Endothelial Cells , Humans , Iron Chelating Agents/pharmacokinetics , Iron Chelating Agents/pharmacology , Liver/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Nanoparticles , Rats , Spleen/metabolism , Tissue Distribution
11.
Clin Exp Pharmacol Physiol ; 45(2): 213-216, 2018 02.
Article in English | MEDLINE | ID: mdl-29205450

ABSTRACT

We evaluated the role of deferasirox therapeutic drug monitoring in order to avoid toxicity or treatment failure. Plasma concentrations, measured between two consecutive liver iron determinations, were determined at the end of dosing interval. Fifty-four ß-thalassemic adult patients were enrolled: 50% were males; median age was 32.3 years (IQR 19.1-41.7 years) and median body mass index was 22.25 kg/m2 (IQR 20.24-23.75 kg/m2 ). The mean deferasirox dose was 28.6 ± 6.3 mg/kg/d and mean plasma concentration was 17.3 ± 16.8 µg/mL. Drug levels showed lower results in males. Deferasirox concentration was significantly correlated with serum creatinine levels (P = .01) and serum ferritin (P < .0001). The assessment of deferasirox therapeutic drug monitoring could help clinicians to predict patient responses and to optimize the therapy.


Subject(s)
Deferasirox/pharmacokinetics , Deferasirox/therapeutic use , Iron Overload/drug therapy , beta-Thalassemia/drug therapy , Adult , Cohort Studies , Dose-Response Relationship, Drug , Female , Humans , Iron Chelating Agents/pharmacokinetics , Iron Chelating Agents/therapeutic use , Male , Young Adult
12.
Am J Hematol ; 92(12): 1356-1361, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28940308

ABSTRACT

Our phase I, open-label, multi-center, dose-escalation study evaluated the pharmacokinetics (PK) of SP-420, a tridentate oral iron chelating agent of the desferrithiocin class, in patients with transfusion dependent ß-thalassemia. SP-420 was administered as a single dose of 1.5 (n = 3), 3 (n = 3), 6 (n = 3), 12 (n = 3), and 24 (n = 6) mg/kg or as a twice-daily dose of 9 mg/kg (n = 6) over 14-28 days. There was a near dose-linear increase in the mean plasma SP-420 concentrations and in the mean values for Cmax and AUC0-τ over the dose range evaluated. The median tmax ranged from 0.5 to 2.25 h and was not dose dependent. The study was prematurely terminated by the sponsor due to renal adverse events (AE) including proteinuria, increase in serum creatinine, and one case of Fanconi syndrome. Other adverse effects included hypersensitivity reactions and gastrointestinal disturbances. Based on current dose administration, the renal AE observed outweighed the possible benefits from chelation therapy. However, additional studies assessing efficacy and safety of lower doses or less frequent dosing of SP-420 over longer durations with close monitoring would be necessary to better explain the findings of our study and characterize the safety of the study drug.


Subject(s)
Cyclohexanones/pharmacokinetics , Dihydropyridines/adverse effects , Iron Chelating Agents/adverse effects , Iron Chelating Agents/pharmacokinetics , Thiazoles/adverse effects , Thiazoles/pharmacokinetics , beta-Thalassemia/therapy , Adolescent , Adult , Blood Transfusion , Cyclohexanones/adverse effects , Cyclohexanones/therapeutic use , Dihydropyridines/therapeutic use , Dose-Response Relationship, Drug , Humans , Iron Chelating Agents/administration & dosage , Kidney Diseases/chemically induced , Middle Aged , Siderophores/therapeutic use , Siderophores/toxicity , Thiazoles/therapeutic use , Young Adult , beta-Thalassemia/complications , beta-Thalassemia/drug therapy
13.
Plant Physiol Biochem ; 118: 579-588, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28787660

ABSTRACT

The growing concern over the environmental risk of synthetic chelate application promotes the search for alternatives in Fe fertilization, such as biodegradable chelating agents and natural complexing agents. In this work, plant responses to the application of several Fe treatments (chelates and complexes) was analyzed to study their potential use in Fe fertilization under calcareous conditions. Thus, the root ferric chelate reductase (FCR) activity of soybean (Glycine max cv. Klaxon) plants was determined, and the effectiveness of the Fe chelates and complexes assessed in a pot experiment, by SPAD and fluorescence induction measurements, and the determination of Fe distribution in plant and soil. Additionally, 57Fe Mössbauer spectroscopy was conducted to identify the Fe forms present in the soybean roots. The highest FCR activity was observed for the chelates EDDS/Fe3+ and IDHA/Fe3+; while no activity was observed when using complexes as Fe substrates. In contrast to the FCR data, the pot experiment confirmed that the o,oEDDHA/Fe3+ is the most effective treatment, and the complexes LS/Fe3+ and GA/Fe3+ are able to alleviate Fe chlorosis, also indicated by SPAD data and the maximal quantum efficiency of photosystem II reaction centers as vitality parameters, and the enhanced plant uptake of Fe from natural sources.


Subject(s)
Glycine max/metabolism , Iron Chelating Agents/pharmacology , Iron , Oxidoreductases/metabolism , Plant Proteins/metabolism , Plant Roots/metabolism , Iron/metabolism , Iron/pharmacology , Iron Chelating Agents/pharmacokinetics
14.
J Cereb Blood Flow Metab ; 37(9): 3110-3123, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28534662

ABSTRACT

Iron overload plays a key role in the secondary brain damage that develops after intracerebral hemorrhage (ICH). The significant increase in iron deposition is associated with the generation of reactive oxygen species (ROS), which leads to oxidative brain damage. In this study, we examined the protective effects of VK-28, a brain-permeable iron chelator, against hemoglobin toxicity in an ex vivo organotypic hippocampal slice culture (OHSC) model and in middle-aged mice subjected to an in vivo, collagenase-induced ICH model. We found that the effects of VK-28 were similar to those of deferoxamine (DFX), a well-studied iron chelator. Both decreased cell death and ROS production in OHSCs and in vivo, decreased iron-deposition and microglial activation around hematoma in vivo, and improved neurologic function. Moreover, compared with DFX, VK-28 polarized microglia to an M2-like phenotype, reduced brain water content, deceased white matter injury, improved neurobehavioral performance, and reduced overall death rate after ICH. The protection of VK-28 was confirmed in a blood-injection ICH model and in aged-male and young female mice. Our findings indicate that VK-28 is protective against iron toxicity after ICH and that, at the dosage tested, it has better efficacy and less toxicity than DFX does.


Subject(s)
Aging/metabolism , Cerebral Hemorrhage/drug therapy , Hippocampus/drug effects , Iron Chelating Agents/therapeutic use , Iron/metabolism , Neuroprotective Agents/therapeutic use , Piperazines/therapeutic use , Quinolines/therapeutic use , Aging/pathology , Animals , Cerebral Hemorrhage/diagnostic imaging , Cerebral Hemorrhage/metabolism , Disease Models, Animal , Female , Hemoglobins/metabolism , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Hippocampus/pathology , In Vitro Techniques , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/pharmacokinetics , Male , Mice, Inbred C57BL , Microscopy, Fluorescence , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Piperazines/administration & dosage , Piperazines/pharmacokinetics , Quinolines/administration & dosage , Quinolines/pharmacokinetics
15.
Pharmacogenomics ; 18(6): 539-554, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28346059

ABSTRACT

AIM: We aimed to evaluate the influence of genetic polymorphisms involved in deferasirox metabolism and transport on its pharmacokinetics and treatment toxicity, in a cohort of ß-thalassaemic children. PATIENTS & METHODS: Drug plasma concentrations were measured by a HPLC-UV method. Allelic discrimination for UGT1A1, UGT1A3, CYP1A1, CYP1A2, CYP2D6, MRP2 and BCRP1 polymorphisms was performed by real-time PCR. RESULTS: CYP1A1 rs2606345AA influenced Ctrough (p = 0.001) and t1/2 (p = 0.042), CYP1A1 rs4646903TC/CC (p = 0.005) and BCRP1 rs2231142GA/AA (p = 0.005) influenced Tmax and CYP2D6 rs1135840CG/GG influenced Cmax (p = 0.044). UGT1A1 rs887829TT (p = 0.002) and CYP1A2 rs762551CC (p = 0.019) resulted as predictive factor of ferritin levels and CYP1A1 rs2606345CA/AA (p = 0.021) and CYP1A2 rs762551AC/CC (p = 0.027) of liver iron concentration. CONCLUSION: Our data suggest the usefulness of deferasirox pharmacogenetics in pediatric treatment optimization.


Subject(s)
Benzoates , Iron Chelating Agents , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Triazoles , beta-Thalassemia/drug therapy , Adolescent , Benzoates/blood , Benzoates/therapeutic use , Benzoates/toxicity , Child , Child, Preschool , Cohort Studies , Deferasirox , Female , Humans , Iron Chelating Agents/pharmacokinetics , Iron Chelating Agents/therapeutic use , Iron Chelating Agents/toxicity , Male , Pharmacogenomic Testing , Triazoles/blood , Triazoles/therapeutic use , Triazoles/toxicity , beta-Thalassemia/genetics , beta-Thalassemia/metabolism
16.
Ther Drug Monit ; 39(2): 185-191, 2017 04.
Article in English | MEDLINE | ID: mdl-28141745

ABSTRACT

BACKGROUND: ß-Thalassemia major patients with higher total drug levels [deferasirox (DEFR) plus its iron complex] do not yield better serum ferritin (SF) control. This study aimed to determine the concentrations of DEFR and its iron complex (Fe-[DEFR]2) in thalassemia patients to predict the chelation efficacy in terms of SF and cardiac T2* values. METHODS: Patients' steady-state drug levels at trough (Ctrough) and 2 hours postdose (C2h) were determined. Because iron deposition may cause changes in the hepatic metabolism of amino acids, the concentrations of 40 amino acids in plasma were also assayed at 2 hours postdose. RESULTS: A total of 28 patients either dosing daily or twice daily were recruited. After a 1-month DEFR maintenance therapy, 38.8% and 30% of patients from groups of once-daily and twice-daily, respectively, had a plasma DEFR-iron complex formation ratio higher than 0.05 [High Chelation Ratio, (HCR)]. After a 6-month follow-up, those patients who had a HCR (n = 10) at C2h showed more favorable median changes in SF and cardiac T2* values (-388.0, +10.1) than those with a low DEFR-iron complex formation ratio (Low Chelation Ratio; n = 18; +10.5; +4.5) compared with the baseline. The levels of plasma L-arginine, L-alanine, L-glycine, L-norleucine, and L-serine were significantly lower in patients with the low Chelation Ratio condition than the levels in HCR patients. CONCLUSIONS: This therapeutic drug monitoring study revealed that a DEFR-iron complex formation ratio at C2h might be an applicable indicator of the efficacy of long-term DEFR iron chelation therapy. A better iron-control response to DEFR was observed in the patients with HCRs. The trends for the ratio might have value in dose-setting and need to be validated in a larger cohort.


Subject(s)
Benzoates/administration & dosage , Benzoates/blood , Iron Chelating Agents/administration & dosage , Iron/blood , Triazoles/administration & dosage , Triazoles/blood , beta-Thalassemia/blood , beta-Thalassemia/drug therapy , Adolescent , Adult , Amino Acids/blood , Benzoates/pharmacokinetics , Deferasirox , Drug Monitoring/methods , Female , Humans , Iron Chelating Agents/pharmacokinetics , Liver/metabolism , Male , Triazoles/pharmacokinetics , Young Adult
17.
Biomacromolecules ; 18(2): 461-474, 2017 02 13.
Article in English | MEDLINE | ID: mdl-27989126

ABSTRACT

Iron accumulation in substantia nigra pars compacta (SNpc) has been proved to be a prominent pathophysiological feature of Parkinson's diseases (PD), which can induce the death of dopaminergic (DA) neurons, up-regulation of reactive oxygen species (ROS), and further loss of motor control. In recent years, iron chelation therapy has been demonstrated to be an effective treatment for PD, which has shown significant improvements in clinical trials. However, the current iron chelators are suboptimal due to their short circulation time, side effects, and lack of proper protection from chelation with ions in blood circulation. In this work, we designed and constructed iron chelation therapeutic nanoparticles protected by a zwitterionic poly(2-methacryloyloxyethyl phosphorylcholine) (PMPC) to delay the saturation of iron chelators in blood circulation and prolong the in vivo lifetime, with HIV-1 trans-activating transcriptor (TAT) served as a shuttle to enhance the blood-brain barrier (BBB) permeability. We explored and investigated whether the Parkinsonian neurodegeneration and the corresponding symptoms in behaviors and physiologies could be prevented or reversed both in vitro and in vivo. The results demonstrated that iron chelator loaded therapeutic nanoparticles could reverse functional deficits in Parkinsonian mice not only physiologically but also behaviorally. On the contrary, both untreated PD mice and non-TAT anchored nanoparticle treated PD mice showed similar loss in DA neurons and difficulties in behaviors. Therefore, with protection of zwitterionic polymer and prolonged in vivo lifetime, iron chelator loaded nanoparticles with delayed saturation provide a PD phenotype reversion therapy and significantly improve the living quality of the Parkinsonian mice.


Subject(s)
Iron Chelating Agents/administration & dosage , Iron/metabolism , Nanoparticles/administration & dosage , Neuroblastoma/drug therapy , Parkinson Disease/drug therapy , Animals , Behavior, Animal/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/metabolism , Cell Survival/drug effects , Disease Models, Animal , Humans , Iron Chelating Agents/pharmacokinetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Nanoparticles/chemistry , Neuroblastoma/metabolism , Parkinson Disease/metabolism , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/chemistry , Polymethacrylic Acids/chemistry , Tissue Distribution , Tumor Cells, Cultured
18.
Br J Clin Pharmacol ; 83(3): 593-602, 2017 03.
Article in English | MEDLINE | ID: mdl-27641003

ABSTRACT

AIMS: Despite long clinical experience with deferiprone, there is limited information on its pharmacokinetics in children aged <6 years. Here we assess the impact of developmental growth on the pharmacokinetics of deferiprone in this population using a population approach. Based on pharmacokinetic bridging concepts, we also evaluate whether the recommended doses yield appropriate systemic exposure in this group of patients. METHODS: Data from a study in which 18 paediatric patients were enrolled were available for the purposes of this analysis. Patients were randomised to three deferiprone dose levels (8.3, 16.7 and 33.3 mg kg-1 ). Blood samples were collected according to an optimised sampling scheme in which each patient contributed to a maximum of five samples. A population pharmacokinetic model was developed using NONMEM v.7.2. Model selection criteria were based on graphical and statistical summaries. RESULTS: A one-compartment model with first-order absorption and first-order elimination best described the pharmacokinetics of deferiprone. Drug disposition parameters were affected by body weight, with both clearance and volume increasing allometrically with size. Simulation scenarios show that comparable systemic exposure (AUC) is achieved in children and adults after similar dose levels in mg kg-1 , with median (5-95th quantiles) AUC values, respectively, of 340.6 (223.2-520.0) µmol l-1  h and 318.5 (200.4-499.0) µmol l-1  h at 75 mg kg-1 day-1 , and 453.7 (297.3-693.0) µmol l-1  h and 424.2 (266.9-664.0) µmol l-1  h at 100 mg kg-1  day-1 given as three times daily (t.i.d.) doses. CONCLUSIONS: Based on the current findings, a dosing regimen of 25 mg kg-1  t.i.d. is recommended in children aged <6 years, with the possibility of titration up to 33.3 mg kg-1  t.i.d.


Subject(s)
Pyridones/administration & dosage , Pyridones/pharmacokinetics , Child, Preschool , Computer Simulation , Deferiprone , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Infant , Iron Chelating Agents/pharmacokinetics , Male , Models, Biological , Pyridones/blood , Single-Blind Method
19.
Mol Nutr Food Res ; 61(3)2017 03.
Article in English | MEDLINE | ID: mdl-27794191

ABSTRACT

SCOPE: Excess free-iron is detrimental to health through its ability to participate in free radical generation and amplification of oncogenic pathways. The study aims were to identify polyphenols with iron-chelating potential. METHODS AND RESULTS: Of four polyphenols tested quercetin demonstrated potent iron binding with the physiological outcome dictated by the location of interaction. In the presence of extracellular iron and quercetin, ferritin expression and cellular iron concentrations decreased suggesting the resulting quercetin-iron complex is not internalised. However, in the relative absence of extracellular iron, quercetin becomes internalised and complexes with both intracellular iron, and iron which subsequently becomes absorbed as indicated by increased cellular 59 Fe post pre-culture with quercetin. This increased intracellular iron complexed to quercetin does not associate with the labile iron pool and cells behave as though they are iron deficient (increased transferrin receptor-1 and iron regulatory protein-2 expression and low ferritin expression). Additionally, a suppression in reactive oxygen species was observed. CONCLUSION: Quercetin, an exogenous iron chelator, is able to render the cell functionally iron-deficient which not only provides a therapeutic platform for chelating excess free luminal iron but also may be of use in limiting processes such as cancer-cell growth, inflammation and bacterial infections, which all require iron.


Subject(s)
Iron Chelating Agents/pharmacology , Iron/metabolism , Polyphenols/pharmacology , Quercetin/pharmacology , Reactive Oxygen Species/metabolism , Anthocyanins/pharmacokinetics , Anthocyanins/pharmacology , Antigens, CD/metabolism , Antioxidants/pharmacology , Biological Transport , Catechin/pharmacokinetics , Catechin/pharmacology , Cell Line, Tumor , Ferritins/metabolism , Glucosides/pharmacokinetics , Glucosides/pharmacology , Glutathione Peroxidase/metabolism , Humans , Iron/pharmacokinetics , Iron Chelating Agents/pharmacokinetics , Iron Regulatory Protein 2/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase , Polyphenols/pharmacokinetics , Quercetin/pharmacokinetics , Receptors, Transferrin/metabolism , Rutin/pharmacokinetics , Rutin/pharmacology
20.
J Pharm Pharmacol ; 69(5): 525-528, 2017 May.
Article in English | MEDLINE | ID: mdl-27230486

ABSTRACT

OBJECTIVES: Iron chelation in the transfusion-dependent anaemias management is essential to prevent end-organ damage and to improve survival. Deferasirox is a once-daily orally active tridentate selective iron chelator which pharmacokinetic disposition could influence treatment efficacy and toxicity. Therapeutic drug monitoring is an important tool for optimizing drug utilization and doses. METHODS: A fully validated chromatographic method was used to quantify deferasirox concentration in plasma collected from paediatric patients with ß-thalassaemia. Samples obtained after 5 days of washout or in naïve patients before and after 2, 4, 6 and 24 h drug administration were evaluated. KEY FINDINGS: Associations between variables were tested using the Pearson test. Twenty paediatric patients were enrolled; they were mainly men (13.65%), with median age of 6.35 years and body mass index of 15.45 kg/m2 . Concerning pharmacokinetic parameters, a higher interindividual variability was shown. A positive, but not significant, correlation (r = 0.363; P = 0.115) was found between deferasirox area under the concentration curve over 24 h (AUC) and drug dose. CONCLUSIONS: Monitoring plasma deferasirox concentrations appears beneficial for guiding appropriate patient treatment, enhancing effectiveness and minimizing toxicity.


Subject(s)
Benzoates/pharmacokinetics , Benzoates/therapeutic use , Iron Chelating Agents/pharmacokinetics , Iron Chelating Agents/therapeutic use , Triazoles/pharmacokinetics , Triazoles/therapeutic use , beta-Thalassemia/drug therapy , beta-Thalassemia/metabolism , Adolescent , Child , Child, Preschool , Cohort Studies , Deferasirox , Female , Humans , Iron/metabolism , Iron Overload/drug therapy , Iron Overload/metabolism , Male , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...