Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
J Biomol Screen ; 18(4): 407-19, 2013 Apr.
Article de Anglais | MEDLINE | ID: mdl-23150017

RÉSUMÉ

Translation initiation is a fine-tuned process that plays a critical role in tumorigenesis. The use of small molecules that modulate mRNA translation provides tool compounds to explore the mechanism of translational initiation and to further validate protein synthesis as a potential pharmaceutical target for cancer therapeutics. This report describes the development and use of a click beetle, dual luciferase cell-based assay multiplexed with a measure of compound toxicity using resazurin to evaluate the differential effect of natural products on cap-dependent or internal ribosome entry site (IRES)-mediated translation initiation and cell viability. This screen identified a series of cardiac glycosides as inhibitors of IRES-mediated translation using, in particular, the oncogene mRNA c-Myc IRES. Treatment of c-Myc-dependent cancer cells with these compounds showed a decrease in c-Myc protein associated with a significant modulation of cell viability. These findings suggest that inhibition of IRES-mediated translation initiation may be a strategy to inhibit c-Myc-driven tumorigenesis.


Sujet(s)
Glucosides cardiotoniques/analyse , Glucosides cardiotoniques/pharmacologie , Évaluation préclinique de médicament , Biosynthèse des protéines/effets des médicaments et des substances chimiques , Inhibiteurs de la synthèse protéique/pharmacologie , Protéines proto-oncogènes c-myc/métabolisme , Ribosomes/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Séquence nucléotidique , Dosage biologique , Glucosides cardiotoniques/composition chimique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Cymarine/composition chimique , Cymarine/pharmacologie , Altération de l'ADN , Gènes rapporteurs , Cellules HEK293 , Humains , Concentration inhibitrice 50 , Inhibiteurs de la synthèse protéique/analyse , Inhibiteurs de la synthèse protéique/composition chimique , Protéines proto-oncogènes c-myc/antagonistes et inhibiteurs , Ribosomes/effets des médicaments et des substances chimiques , Facteur de croissance endothéliale vasculaire de type A/métabolisme
2.
Exp Hematol ; 40(10): 820-7, 2012 Oct.
Article de Anglais | MEDLINE | ID: mdl-22713799

RÉSUMÉ

Iron chelation is essential to patients on chronic blood transfusions to prevent toxicity from iron overload and remove excess iron. Deferasirox (DFX) is the most commonly used iron chelator in the United States; however, some patients are relatively refractory to DFX therapy. We postulated that vitamin C supplementation would improve the availability of transfusional iron to DFX treatment by promoting iron's redox cycling, increasing its soluble ferrous form and promoting its release from reticuloendothelial cells. Osteogenic dystrophy rats (n = 54) were given iron dextran injections for 10 weeks. Cardiac and liver iron levels were measured after iron loading (n = 18), 12 weeks of sham chelation (n = 18), and 12 weeks of DFX chelation (n = 18) at 75 mg/kg/day. Ascorbate supplementation of 150 ppm, 900 ppm, and 2250 ppm was used in the chow to mimic a broad range of ascorbate status; plasma ascorbate levels were 5.4 ± 1.9, 8.2 ± 1.4, 23.6 ± 9.8 µM, respectively (p < 0.0001). The most severe ascorbate deficiency produced reticuloenthelial retention, lowering total hepatic iron by 29% at the end of iron loading (p < 0.05) and limiting iron redistribution from cardiac and hepatic macrophages during 12 weeks of sham chelation. Most importantly, ascorbate supplementation at 2250 ppm improved DFX efficiency, allowing DFX to remove 21% more hepatic iron than ascorbate supplementation with 900 ppm or 150 ppm (p < 0.05). We conclude that vitamin C status modulates the release of iron from the reticuloendothelial system and correlates positively with DFX chelation efficiency. Our findings suggest that ascorbate status should be probed in patients with unsatisfactory response to DFX.


Sujet(s)
Carence en acide ascorbique , Acide ascorbique/sang , Benzoates/pharmacologie , Agents chélateurs du fer/pharmacologie , Surcharge en fer , Fer/sang , Système phagocytaire mononucléé/métabolisme , Triazoles/pharmacologie , Animaux , Carence en acide ascorbique/sang , Carence en acide ascorbique/traitement médicamenteux , Déférasirox , Cochons d'Inde , Humains , Surcharge en fer/sang , Surcharge en fer/traitement médicamenteux , Foie/métabolisme , Foie/anatomopathologie , Système phagocytaire mononucléé/anatomopathologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Rats , Souches mutantes de rat
4.
Eur J Haematol ; 82(6): 454-7, 2009 Jun.
Article de Anglais | MEDLINE | ID: mdl-19191863

RÉSUMÉ

This subgroup analysis evaluated the effect of once-daily oral deferasirox on labile plasma iron (LPI) levels in patients from the prospective, 1-yr, multicentre ESCALATOR study. Mean baseline liver iron concentration and median serum ferritin levels were 28.6 +/- 10.3 mg Fe/g dry weight and 6334 ng/mL respectively, indicating high iron burden despite prior chelation therapy. Baseline LPI levels (0.98 +/- 0.82 micromol/L) decreased significantly to 0.12 +/- 0.16 micromol/L, 2 h after first deferasirox dose (P = 0.0006). Reductions from pre- to post-deferasirox administration were also observed at all other time points. Compared to baseline, there was a significant reduction in preadministration LPI that reached the normal range at week 4 and throughout the remainder of the study (P < or = 0.02). Pharmacokinetic analysis demonstrated an inverse relationship between preadministration LPI levels and trough deferasirox plasma concentrations. Once-daily dosing with deferasirox > or =20 mg/kg/d provided sustained reduction in LPI levels in these heavily iron-overloaded patients, suggesting 24-h protection from LPI. Deferasirox may therefore reduce unregulated tissue iron loading and prevent further end-organ damage.


Sujet(s)
Benzoates/administration et posologie , Agents chélateurs du fer/administration et posologie , Surcharge en fer/traitement médicamenteux , Fer/sang , Triazoles/administration et posologie , bêta-Thalassémie/traitement médicamenteux , Adolescent , Adulte , Benzoates/pharmacocinétique , Enfant , Déférasirox , Femelle , Ferritines/sang , Humains , Fer/analyse , Agents chélateurs du fer/pharmacocinétique , Foie/composition chimique , Mâle , Résultat thérapeutique , Triazoles/pharmacocinétique , Jeune adulte
5.
Exp Biol Med (Maywood) ; 234(5): 492-503, 2009 May.
Article de Anglais | MEDLINE | ID: mdl-19234060

RÉSUMÉ

Mutations in hemojuvelin (HJV) cause severe juvenile hemochromatosis, characterized by iron loading of the heart, liver, and pancreas. Knockout (KO) mice lacking HJV (Hjv-/-) spontaneously load with dietary iron and, therefore, present a model for hereditary hemochromatosis (HH). In HH, iron chelation may be considered in noncandidates for phlebotomy. We examined the effects of deferasirox, an oral chelator, in Hjv-/- mice. Hepatic, cardiac, splenic, and pancreatic iron were determined by measuring elemental iron and scoring histological sections. Heart and liver iron levels were also determined repeatedly by quantitative R2* magnetic resonance imaging (MRI). The time course of iron loading without intervention was followed from Week 8 of age (study start) to Week 20, when once-daily (5x/week) deferasirox was administered, to Week 28. At 8 weeks, liver iron of KO mice was already markedly elevated versus wild-type mice (P<0.001) and reached a plateau around Week 14. In contrast, Week 8 cardiac and pancreatic iron levels were similar in both KO and wild-type mice and, compared with the liver, showed a delayed but massive iron loading up to Week 20. Contrary to the liver, heart, and pancreas, the KO mice spleen had lower iron content versus wild-type mice. In Hjv-/- mice, liver and heart iron burden was effectively reduced with deferasirox 100 mg/kg (P<0.05). Although deferasirox was less efficacious at this dose in the pancreas, over the observed time period, a clear trend toward reduced organ iron load was noted. There was no noticeable effect of deferasirox upon splenic iron in Hjv-/- mice. Quantitative R2* MRI demonstrated the ability to assess iron concentrations in the liver and myocardial muscle accurately and repetitively. Hepatic (R=0.86; P=3.2*10(-12)) and delayed myocardial (R=0.81; P=2.9*10(-10)) iron accumulation could be followed noninvasively with high agreement to invasive methods.


Sujet(s)
Benzoates/pharmacologie , Hémochromatose/traitement médicamenteux , Agents chélateurs du fer/pharmacologie , Fer/métabolisme , Triazoles/pharmacologie , Animaux , Benzoates/usage thérapeutique , Déférasirox , Modèles animaux de maladie humaine , Évaluation préclinique de médicament , Protéines liées au GPI , Hémochromatose/génétique , Protéine de l'hémochromatose , Humains , Agents chélateurs du fer/usage thérapeutique , Imagerie par résonance magnétique , Protéines membranaires/génétique , Souris , Souris knockout , Spécificité d'organe , Triazoles/usage thérapeutique
6.
Acta Haematol ; 120(2): 123-8, 2008.
Article de Anglais | MEDLINE | ID: mdl-19018129

RÉSUMÉ

INTRODUCTION: Combined therapy with deferoxamine (DFO) and deferasirox (DFX) may be performed empirically when DFX monotherapy fails. Given the lack of published data on this therapy, the study goal was to assess the safety and efficacy of combined DFO/DFX therapy in a gerbil model. METHODS: Thirty-two female Mongolian gerbils 8-10 weeks old were divided into 4 groups (sham chelated, DFO, DFX, DFO/DFX). Each received 10 weekly injections of 200 mg/kg iron dextran prior to initiation of 12 weeks of chelation. Experimental endpoints were heart and liver weights, iron concentration and histology. RESULTS: In the heart, there was no significant difference among the treatment groups for wet-to-dry ratio, iron concentration and iron content. DFX-treated animals exhibited lower organ weights relative to sham-chelated animals (less iron-mediated hypertrophy). DFO-treated organs did not differ from sham-chelated organs in any aspects. DFX significantly cleared hepatic iron. No additive effects were observed in the organs of DFO/DFX-treated animals. CONCLUSIONS: Combined DFO/DFX therapy produced no detectable additive effect above DFX monotherapy in either the liver or heart, suggesting competition with spontaneous iron elimination mechanisms for chelatable iron. Combined therapy was well tolerated, but its efficacy could not be proven due to limitations in the animal model.


Sujet(s)
Benzoates/administration et posologie , Déferoxamine/administration et posologie , Surcharge en fer/traitement médicamenteux , Triazoles/administration et posologie , Animaux , Déférasirox , Modèles animaux de maladie humaine , Association de médicaments , Effets secondaires indésirables des médicaments , Gerbillinae , Coeur , Foie , Taille d'organe , Résultat thérapeutique
7.
Drug Metab Dispos ; 36(12): 2523-38, 2008 Dec.
Article de Anglais | MEDLINE | ID: mdl-18775980

RÉSUMÉ

Deferasirox (Exjade, ICL670, CGP72670) is an iron-chelating drug for p.o. treatment of transfusional iron overload in patients with beta-thalassemia or sickle cell disease. The pharmacokinetics and disposition of deferasirox were investigated in rats. The animals received single intravenous (10 mg/kg) or p.o. (10 or 100 mg/kg) doses of 14C-radiolabeled deferasirox. Biological samples were analyzed for radioactivity (liquid scintillation counting, quantitative whole-body autoradioluminography), for deferasirox and its iron complex [high-performance liquid chromatography (HPLC)/UV], and for metabolites (HPLC with radiodetection, liquid chromatography/mass spectrometry, 1H and 13C NMR, and two-dimensional NMR techniques). At least 75% of p.o.-dosed deferasirox was absorbed. The p.o. bioavailability was 26% at the 10 mg/kg dose and showed an overproportional increase at the 100 mg/kg dose, probably because of saturation of elimination processes. Deferasirox-related radioactivity was distributed mainly to blood, excretory organs, and gastrointestinal tract. Enterohepatic recirculation of deferasirox was observed. No retention occurred in any tissue. The placental barrier was passed to a low extent. Approximately 3% of the dose was transferred into the breast milk. Excretion of deferasirox and metabolites was rapid and complete within 7 days. Key clearance processes were hepatic metabolism and biliary elimination via multidrug resistance protein 2. Deferasirox, iron complex, and metabolites were excreted largely via bile and feces (total > or = 90%). Metabolism included glucuronidation at the carboxylate group (acyl glucuronide M3) and at phenolic hydroxy groups, as well as, to a lower degree, cytochrome P450-catalyzed hydroxylations. Two hydroxylated metabolites (M1 and M2) were administered to rats and were shown not to contribute substantially to iron elimination in vivo.


Sujet(s)
Benzoates/métabolisme , Benzoates/pharmacocinétique , Fer/métabolisme , Triazoles/métabolisme , Triazoles/pharmacocinétique , Transporteurs ABC/génétique , Transporteurs ABC/métabolisme , Animaux , Animaux allaités/métabolisme , Benzoates/administration et posologie , Bile/composition chimique , Analyse chimique du sang , Déférasirox , Circulation entérohépatique , Fèces/composition chimique , Femelle , Foetus/métabolisme , Fer/analyse , Agents chélateurs du fer/administration et posologie , Agents chélateurs du fer/métabolisme , Agents chélateurs du fer/pharmacologie , Spectroscopie par résonance magnétique , Mâle , Spectrométrie de masse , Lait humain/composition chimique , Lait humain/métabolisme , Modèles biologiques , Structure moléculaire , Placenta/métabolisme , Grossesse , Rats , Rat Long-Evans , Rats transgéniques , Rat Wistar , Distribution tissulaire , Triazoles/administration et posologie , Urine/composition chimique
8.
Magn Reson Med ; 60(1): 82-9, 2008 Jul.
Article de Anglais | MEDLINE | ID: mdl-18581418

RÉSUMÉ

MRI is gaining increasing importance for the noninvasive quantification of organ iron burden. Since transverse relaxation rates depend on iron distribution as well as iron concentration, physiologic and pharmacologic processes that alter iron distribution could change MRI calibration curves. This article compares the effect of three iron chelators, deferoxamine, deferiprone, and deferasirox, on R1 and R2 calibration curves according to two iron loading and chelation strategies. Thirty-three Mongolian gerbils underwent iron loading (iron dextran 500 mg/kg/wk) for 3 weeks followed by 4 weeks of chelation. An additional 56 animals received less aggressive loading (200 mg/kg/week) for 10 weeks, followed by 12 weeks of chelation. R1 and R2 calibration curves were compared to results from 23 iron-loaded animals that had not received chelation. Acute iron loading and chelation-biased R1 and R2 from the unchelated reference calibration curves but chelator-specific changes were not observed, suggesting physiologic rather than pharmacologic differences in iron distribution. Long-term chelation deferiprone treatment increased liver R1 50% (P < 0.01), while long-term deferasirox lowered liver R2 30.9% (P < 0.0001). The relationship between R1 and R2 and organ iron concentration may depend on the acuity of iron loading and unloading as well as the iron chelator administered.


Sujet(s)
Fer/métabolisme , Foie/composition chimique , Imagerie par résonance magnétique , Myocarde/composition chimique , Animaux , Benzoates/pharmacologie , Déférasirox , Défériprone , Déferoxamine/pharmacologie , Gerbillinae , Agents chélateurs du fer/pharmacologie , Pyridones/pharmacologie , Sidérophores/pharmacologie , Distribution tissulaire , Triazoles/pharmacologie
9.
Curr Opin Chem Biol ; 11(4): 419-23, 2007 Aug.
Article de Anglais | MEDLINE | ID: mdl-17644021

RÉSUMÉ

Orally bioavailable chelators for transfusional iron overload have been sought since the introduction of deferoxamine (Desferal) in 1962. Despite tremendous efforts, to date, only deferiprone (Ferriprox) and deferasirox (Exjade) have successfully reached the market, reflecting the difficulty to combine oral activity and safety. Owing to the risk of failure, few new oral chelators can be expected in the future for the treatment of transfusional iron overload. As iron is involved in many disease processes, deferiprone and deferasirox have been proposed to be potentially useful in a variety of indications not characterized by general iron overload. Although it may be possible to obtain clinical benefit from current compounds, more selective chelators tailored to the particular target are needed for successful intervention in these indications.


Sujet(s)
Agents chélateurs du fer/pharmacologie , Administration par voie orale , Transport biologique , Déferoxamine/pharmacologie , Hémochromatose/traitement médicamenteux , Hémochromatose/génétique , Hémochromatose/métabolisme , Humains , Ions/métabolisme , Agents chélateurs du fer/administration et posologie , Agents chélateurs du fer/composition chimique
10.
Exp Hematol ; 35(7): 1069-73, 2007 Jul.
Article de Anglais | MEDLINE | ID: mdl-17588475

RÉSUMÉ

OBJECTIVE: Despite the availability of deferoxamine chelation therapy for more than 20 years, iron cardiomyopathy remains the leading cause of death in thalassemia major patients. Effective chelation of cardiac iron is difficult; cardiac iron stores respond more slowly to chelation therapy and require a constant gradient of labile iron species between serum and myocytes. We have previously demonstrated the efficacy of once-daily deferasirox in removing previously stored cardiac iron in the gerbil, but changes in cardiac iron were relatively modest compared with hepatic iron. We postulated that daily divided dosing, by sustaining a longer labile iron gradient from myocytes to serum, would produce better cardiac iron chelation than a comparable daily dose. METHODS: Twenty-four 8- to 10-week-old female gerbils underwent iron dextran-loading for 10 weeks, followed by a 1-week iron equilibration period. Animals were divided into three treatment groups of eight animals each and were treated with deferasirox 100 mg/kg/day as a single dose, deferasirox 100 mg/kg/day daily divided dose, or sham chelation for a total of 12 weeks. Following euthanasia, organs were harvested for quantitative iron and tissue histology. RESULTS: Hepatic and cardiac iron contents were not statistically different between the daily single-dose and daily divided-dose groups. However, the ratio of cardiac to hepatic iron content was lower in the divided-dose group (0.78% vs 1.11%, p = 0.0007). CONCLUSION: Daily divided dosing of deferasirox changes the relative cardiac and liver iron chelation profile compared with daily single dosing, trading improvements in cardiac iron elimination for less-effective hepatic chelation.


Sujet(s)
Benzoates/administration et posologie , Cardiomyopathies/traitement médicamenteux , Modèles animaux de maladie humaine , Agents chélateurs du fer/administration et posologie , Surcharge en fer/traitement médicamenteux , Triazoles/administration et posologie , Animaux , Cardiomyopathies/métabolisme , Cardiomyopathies/anatomopathologie , Déférasirox , Calendrier d'administration des médicaments , Femelle , Gerbillinae , Fer/métabolisme , Surcharge en fer/métabolisme , Surcharge en fer/anatomopathologie , Foie/métabolisme , Foie/anatomopathologie , Myocarde/métabolisme , Myocarde/anatomopathologie
11.
Transl Res ; 148(5): 272-80, 2006 Nov.
Article de Anglais | MEDLINE | ID: mdl-17145573

RÉSUMÉ

INTRODUCTION: Deferasirox effectively controls liver iron concentration; however, little is known regarding its ability to remove stored cardiac iron. Deferiprone seems to have increased cardiac efficacy compared with traditional deferoxamine therapy. Therefore, the relative efficacy of deferasirox and deferiprone were compared in removing cardiac iron from iron-loaded gerbils. METHODS: Twenty-nine 8- to 10-week-old female gerbils underwent 10 weekly iron dextran injections of 200 mg/kg/week. Prechelation iron levels were assessed in 5 animals, and the remainder received deferasirox 100 mg/kg/D po QD (n = 8), deferiprone 375 mg/kg/D po divided TID (n = 8), or sham chelation (n = 8), 5 days/week for 12 weeks. RESULTS: Deferasirox reduced cardiac iron content 20.5%. No changes occurred in cardiac weight, myocyte hypertrophy, fibrosis, or weight-to-dry weight ratio. Deferasirox treatment reduced liver iron content 51%. Deferiprone produced comparable reductions in cardiac iron content (18.6% reduction). Deferiprone-treated hearts had greater mass (16.5% increase) and increased myocyte hypertrophy. Deferiprone decreased liver iron content 24.9% but was associated with an increase in liver weight and water content. CONCLUSION: Deferasirox and deferiprone were equally effective in removing stored cardiac iron in a gerbil animal model, but deferasirox removed more hepatic iron for a given cardiac iron burden.


Sujet(s)
Benzoates/usage thérapeutique , Agents chélateurs du fer/usage thérapeutique , Surcharge en fer/traitement médicamenteux , Pyridones/usage thérapeutique , Triazoles/usage thérapeutique , Animaux , Déférasirox , Défériprone , Modèles animaux de maladie humaine , Femelle , Gerbillinae , Coeur/effets des médicaments et des substances chimiques , Fer/métabolisme , Surcharge en fer/anatomopathologie , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Taille d'organe/effets des médicaments et des substances chimiques
12.
Blood ; 108(9): 3195-203, 2006 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-16835377

RÉSUMÉ

Labile iron in hemosiderotic plasma and tissue are sources of iron toxicity. We compared the iron chelators deferoxamine, deferiprone, and deferasirox as scavengers of labile iron in plasma and cardiomyocytes at therapeutic concentrations. This comprised chelation of labile plasma iron (LPI) in samples from thalassemia patients; extraction of total cellular iron; accessing labile iron accumulated in organelles and preventing formation of reactive-oxidant species; and restoring impaired cardiac contractility. Neonatal rat cardiomyocytes were used for monitoring chelator extraction of LCI (labile cell iron) as 59Fe; assessing in situ cell iron chelation by epifluorescence microscope imaging using novel fluorescent sensors for iron and reactive oxygen species (ROS) selectively targeted to organelles, and monitoring contractility by time-lapse microscopy. At plasma concentrations attained therapeutically, all 3 chelators eliminated LPI but the orally active chelators rapidly gained access to the LCI pools of cardiomyocytes, bound labile iron, attenuated ROS formation, extracted accumulated iron, and restored contractility impaired by iron overload. The effect of deferoxamine at therapeutically relevant concentrations was primarily by elimination of LPI. The rapid accessibility of the oral chelators deferasirox and deferiprone to intracellular labile iron compartments renders them potentially efficacious for protection from and possibly reversal of cardiac damage induced by iron overload.


Sujet(s)
Agents chélateurs du fer/pharmacologie , Fer/métabolisme , Myocarde/métabolisme , Animaux , Animaux nouveau-nés , Fluorescéines , Coeur/effets des médicaments et des substances chimiques , Fer/sang , Fer/toxicité , Mitochondries du myocarde/effets des médicaments et des substances chimiques , Mitochondries du myocarde/physiologie , Cellules musculaires/effets des médicaments et des substances chimiques , Cellules musculaires/physiologie , Rats , Espèces réactives de l'oxygène/métabolisme
13.
Eur J Pharmacol ; 541(3): 129-37, 2006 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-16765341

RÉSUMÉ

By comparing the antiproliferative effect of the iron chelators ICL670A and O-trensox in the human hepatoma cell line HUH7 and human hepatocyte cultures, we have shown that ICL670A decreased cell viability, inhibited DNA replication and induced DNA fragmentation more efficiently than O-trensox. O-trensox and ICL670A induced a cell cycle blockade in G0-G1 and S phases respectively. In parallel, ICL670A inhibited polyamine biosynthesis by decreasing ornithine decarboxylase and spermidine/spermine N(1)-acetyltransferase activities. O-trensox increased polyamine biosynthesis and particularly putrescine level by stimulating spermidine-spermine N(1)-acetyltransferase activity which could activate the polyamine retro-conversion pathway. Moreover, the two chelators exhibit some cytotoxic effect in the two culture models; ICL670A was more cytotoxic than O-trensox and higher concentrations of the two chelators were necessary to induce a cytotoxicity in primary cultures versus hepatoma cells. These results suggested that ICL670A has the most efficient antitumoral effect, blocks cell proliferation by a pathway different of O-trensox and may constitute a potential drug for anticancer therapy.


Sujet(s)
Benzoates/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Éthylamines/pharmacologie , Hépatocytes/effets des médicaments et des substances chimiques , Hydroxyquinoléines/pharmacologie , Agents chélateurs du fer/pharmacologie , Triazoles/pharmacologie , Acetyltransferases/génétique , Acetyltransferases/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Polyamines biogènes/métabolisme , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Cycle cellulaire/effets des médicaments et des substances chimiques , Réplication de l'ADN/effets des médicaments et des substances chimiques , Déférasirox , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Humains , Ornithine decarboxylase/génétique , Ornithine decarboxylase/métabolisme , ARN messager/métabolisme
14.
Circulation ; 112(4): 535-43, 2005 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-16027257

RÉSUMÉ

BACKGROUND: Transfusional therapy for thalassemia major and sickle cell disease can lead to iron deposition and damage to the heart, liver, and endocrine organs. Iron causes the MRI parameters T1, T2, and T2* to shorten in these organs, which creates a potential mechanism for iron quantification. However, because of the danger and variability of cardiac biopsy, tissue validation of cardiac iron estimates by MRI has not been performed. In this study, we demonstrate that iron produces similar T1, T2, and T2* changes in the heart and liver using a gerbil iron-overload model. METHODS AND RESULTS: Twelve gerbils underwent iron dextran loading (200 mg . kg(-1) . wk(-1)) from 2 to 14 weeks; 5 age-matched controls were studied as well. Animals had in vivo assessment of cardiac T2* and hepatic T2 and T2* and postmortem assessment of cardiac and hepatic T1 and T2. Relaxation measurements were performed in a clinical 1.5-T magnet and a 60-MHz nuclear magnetic resonance relaxometer. Cardiac and liver iron concentrations rose linearly with administered dose. Cardiac 1/T2*, 1/T2, and 1/T1 rose linearly with cardiac iron concentration. Liver 1/T2*, 1/T2, and 1/T1 also rose linearly, proportional to hepatic iron concentration. Liver and heart calibrations were similar on a dry-weight basis. CONCLUSIONS: MRI measurements of cardiac T2 and T2* can be used to quantify cardiac iron. The similarity of liver and cardiac iron calibration curves in the gerbil suggests that extrapolation of human liver calibration curves to heart may be a rational approximation in humans.


Sujet(s)
Cardiomyopathies/métabolisme , Surcharge en fer/métabolisme , Fer/analyse , Imagerie par résonance magnétique , Myocarde/composition chimique , Thalassémie/thérapie , Animaux , Calibrage , Femelle , Gerbillinae , Humains , Fer/métabolisme , Foie/composition chimique , Reproductibilité des résultats , Spécificité d'espèce , Réaction transfusionnelle
15.
Bioorg Med Chem Lett ; 14(10): 2451-7, 2004 May 17.
Article de Anglais | MEDLINE | ID: mdl-15109631

RÉSUMÉ

Combination of structural elements from a potent Y5 antagonist (2) with thiazole fragments that exhibit weak Y5 affinities followed by lead optimisation led to the discovery of (5,6-dihydro-4H-3-thia-1-aza-benzo[e]azulen-2-yl)-piperidin-4-ylmethyl-amino and (4,5-dihydro-6-oxa-3-thia-1-aza-benzo[e]azulen-2-yl)-piperidin-4-ylmethyl-amino derivatives. Both classes of compounds are capable of delivering potent and selective orally and centrally bioavailable NPY Y5 receptor antagonists.


Sujet(s)
Cycloheptanes/synthèse chimique , Cycloheptanes/pharmacocinétique , Récepteur neuropeptide Y/antagonistes et inhibiteurs , Administration par voie orale , Animaux , Azulènes , Biodisponibilité , Sang , Barrière hémato-encéphalique , Cycloheptanes/pharmacologie , Composés hétérocycliques 3 noyaux/synthèse chimique , Composés hétérocycliques 3 noyaux/pharmacocinétique , Composés hétérocycliques 3 noyaux/pharmacologie , Composés hétérocycliques avec 4 noyaux ou plus/synthèse chimique , Composés hétérocycliques avec 4 noyaux ou plus/pharmacocinétique , Composés hétérocycliques avec 4 noyaux ou plus/pharmacologie , Hypothalamus , Concentration inhibitrice 50 , Rats , Relation structure-activité
16.
Curr Med Chem ; 10(12): 1065-76, 2003 Jun.
Article de Anglais | MEDLINE | ID: mdl-12678677

RÉSUMÉ

Successful treatment of beta-thalassemia requires two key elements: blood transfusion and iron chelation. Regular blood transfusions considerably expand the lifespan of patients, however, without the removal of the consequential accumulation of body iron, few patients live beyond their second decade. In 1963, the introduction of desferrioxamine (DFO), a hexadentate chelator, marked a breakthrough in the treatment of beta-thalassemia. DFO significantly reduces body iron burden and iron-related morbidity and mortality. DFO is still the only drug for general use in the treatment of transfusion dependent iron overload. However, its very short plasma half-life and poor oral activity necessitate special modes of application (subcutaneous or intravenous infusion) which are inconvenient, can cause local reactions and are difficult to be accepted by many patients. Over the past four decades, many different laboratories have invested major efforts in the identification of orally active iron chelators from several hundreds of molecules of synthetic, microbial or plant origin. The discovery of ferrithiocin in 1980, followed by the synthesis of the tridentate chelator desferrithiocin and proof of its oral activity raised a lot of hope. However, the compound proved to be toxic in animals. Over a period of about fifteen years many desferrithiocin derivatives and molecules with broader alterations led to the discovery of numerous new compounds some of which were much better tolerated and were more efficacious than desferrithiocin in animals, however, none was safe enough to proceed to the clinical use. The discovery of a new chemical class of iron chelators: The bis-hydroxyphenyltriazoles re-energized the search for a safe tridentate chelator. The basic structure of this completely new chemical class of iron chelators was discovered by a combination of rational design, intuition and experience. More than forty derivatives of the triazole series were synthesized at Novartis. These compounds were evaluated, together with more than 700 chelators from various chemical classes. Using vigorous selection criteria with a focus on tolerability, the tridentate chelator 4-[(3,5-Bis-(2-hydroxyphenyl)-1,2,4)triazol-1-yl]-benzoic acid (ICL670) emerged as an entity which best combined high oral potency and tolerability in animals. ICL670 is presently being evaluated in the clinic.


Sujet(s)
Benzoates/pharmacologie , Dihydropyridines/pharmacologie , Agents chélateurs du fer/pharmacologie , Thiazoles/pharmacologie , Triazoles/pharmacologie , Animaux , Benzoates/effets indésirables , Benzoates/composition chimique , Benzoates/pharmacocinétique , Benzoates/toxicité , Callithrix , Phénomènes chimiques , Chimie physique , Déférasirox , Dihydropyridines/effets indésirables , Dihydropyridines/composition chimique , Dihydropyridines/pharmacocinétique , Dihydropyridines/toxicité , Chiens , Humains , Fer/composition chimique , Agents chélateurs du fer/effets indésirables , Agents chélateurs du fer/composition chimique , Agents chélateurs du fer/pharmacocinétique , Agents chélateurs du fer/toxicité , Rats , Thiazoles/effets indésirables , Thiazoles/composition chimique , Thiazoles/pharmacocinétique , Thiazoles/toxicité , Triazoles/effets indésirables , Triazoles/composition chimique , Triazoles/pharmacocinétique , Triazoles/toxicité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE