Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 318(1): 424-33, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16614166

ABSTRACT

Antitumor therapy with the anthracycline doxorubicin is limited by a dose-related cardiotoxicity that is aggravated by a concomitant administration of the taxane paclitaxel. Previous limited studies with isolated human heart cytosol showed that paclitaxel was able to stimulate an NADPH-dependent reduction of doxorubicin to its toxic secondary alcohol metabolite doxorubicinol. Here we characterized that 0.25 to 2.5 microM paclitaxel caused allosteric effects that increased doxorubicinol formation in human heart cytosol, whereas 5 to 10 microM paclitaxel decreased doxorubicinol formation. The closely related taxane docetaxel caused similar effects. Basal or taxane-stimulated doxorubicinol formation was blunted by 2,7-difluorospirofluorene-9,5'-imidazolidine-2',4'-dione (AL1576), a specific inhibitor of aldehyde reductases. Doxorubicinol was measured also in the cytosol of human myocardial strips incubated in plasma and exposed to doxorubicin in the absence or presence of paclitaxel or docetaxel and their clinical vehicles Cremophor EL or polysorbate 80. Low concentrations of taxanes stimulated doxorubicinol formation, whereas high concentrations decreased it. Doxorubicinol formation reached its maximum on adding plasma with 6 microM paclitaxel or docetaxel; this corresponded to the partitioning of 1.5 to 2.5 microM taxanes in the cytosol of the strips. Taxane-stimulated doxorubicinol formation was not mediated by vehicles, nor was it caused by increased doxorubicin uptake or de novo protein synthesis; however, doxorubicinol formation was blunted by AL1576. These results show that allosteric interactions with cytoplasmic aldehyde reductases enable paclitaxel or docetaxel to stimulate doxorubicinol formation in human heart. This information serves metabolic insights into the risk of cardiotoxicity induced by doxorubicin-taxane therapies.


Subject(s)
Doxorubicin/analogs & derivatives , Myocardium/metabolism , Paclitaxel/toxicity , Taxoids/toxicity , Aged , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Docetaxel , Dose-Response Relationship, Drug , Doxorubicin/metabolism , Doxorubicin/toxicity , Female , Humans , In Vitro Techniques , Male , Middle Aged , Paclitaxel/metabolism , Taxoids/metabolism
2.
J Biol Chem ; 281(16): 10990-1001, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16423826

ABSTRACT

One-electron quinone reduction and two-electron carbonyl reduction convert the anticancer anthracycline doxorubicin to reactive oxygen species (ROS) or a secondary alcohol metabolite that contributes to inducing a severe form of cardiotoxicity. The closely related analogue epirubicin induces less cardiotoxicity, but the determinants of its different behavior have not been elucidated. We developed a translational model of the human heart and characterized whether epirubicin exhibited a defective conversion to ROS and secondary alcohol metabolites. Small myocardial samples from cardiac surgery patients were reconstituted in plasma that contained clinically relevant concentrations of doxorubicin or epirubicin. In this model only doxorubicin formed ROS, as detected by fluorescent probes or aconitase inactivation. Experiments with cell-free systems and confocal laser scanning microscopy studies of H9c2 cardiomyocytes suggested that epirubicin could not form ROS because of its protonation-dependent sequestration in cytoplasmic acidic organelles and the consequent limited localization to mitochondrial one-electron quinone reductases. Accordingly, blocking the protonation-sequestration mechanism with the vacuolar H+-ATPase inhibitor bafilomycin A1 relocalized epirubicin to mitochondria and increased its conversion to ROS in human myocardial samples. Epirubicin also formed approximately 60% less alcohol metabolites than doxorubicin, but this was caused primarily by its higher Km and lower Vmax values for two-electron carbonyl reduction by aldo/keto-reductases of human cardiac cytosol. Thus, vesicular sequestration and impaired efficiency of electron addition have separate roles in determining a defective bioactivation of epirubicin to ROS or secondary alcohol metabolites in the human heart. These results uncover the molecular determinants of the reduced cardiotoxicity of epirubicin and serve mechanism-based guidelines to improving antitumor therapies.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Epirubicin/pharmacology , Aconitate Hydratase/metabolism , Adenosine Triphosphatases/chemistry , Aged , Antineoplastic Agents/pharmacology , Area Under Curve , Cell Line, Tumor , Cell Membrane/metabolism , Cell-Free System , Cells, Cultured , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Electrons , Epirubicin/chemistry , Female , Fluorescent Dyes/pharmacology , Humans , Hydrogen Peroxide/chemistry , Inhibitory Concentration 50 , Kinetics , Macrolides/chemistry , Male , Microscopy, Confocal , Microscopy, Fluorescence , Middle Aged , Mitochondria/metabolism , Models, Chemical , Myocardium/metabolism , Myocardium/pathology , Oxidation-Reduction , Oxygen/metabolism , Protein Biosynthesis , Protons , Reactive Oxygen Species , Time Factors
3.
Chem Res Toxicol ; 15(9): 1179-89, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12230412

ABSTRACT

Lipid peroxidation has been proposed to mediate cardiotoxicity induced by doxorubicin (DOX) and other anticancer anthracyclines; however, there have been reports showing that DOX can also inhibit lipid peroxidation. Here we characterized the effects of DOX on the oxo-ferryl moiety [Fe(IV)=O, Mb(IV)] of H(2)O(2)-activated myoglobin, a lipid oxidant likely formed in the heart during treatment with DOX. Mb(IV) was formed in vitro by reacting 100 microM H(2)O(2) with 50 microM horse heart metmyoglobin (Mb(III)). Spectral studies showed that DOX reduced Mb(IV) to Mb(III), half-maximal regeneration of Mb(III) occurring at approximately 18 microM DOX. Comparisons between DOX, its aglycone doxorubicinone, and other approved or investigational anthracyclines or model compounds (daunorubicin, idarubicin, aclarubicin, and naphthazarin), showed that DOX reduced Mb(IV) through the hydroquinone moiety of its tetracyclic ring. DOX inhibited Mb(IV)-dependent peroxidation of arachidonic acid, suppressing the formation of thiobarbituric acid-reactive substances with an IC(50) of approximately 18 microM. Lipid peroxidation was inhibited also by the hydroquinone-containing daunorubicin and idarubicin but not by the hydroquinone-deficient aclarubicin; moreover, neither simple hydroquinone nor other known Mb(IV) reductants (ascorbate, glutathione, and ergothioneine) reached measurable IC(50)s in a micromolar range. DOX-dependent inhibition of lipid peroxidation correlated with its ability to reduce Mb(IV) to Mb(III) in competition with arachidonic acid (r = 0.83, P = 0.029); it did not correlate with its ability to scavenge other free radical species [like e.g., peroxyl radicals generated through the thermal decomposition of 2,2'-azo-bis(2-amidinopropane)]. DOX reduced Mb(IV) and inhibited lipid peroxidation also when H(2)O(2), Mb(III) and arachidonic acid were reacted in cytosol of human myocardial biopsies, a model developed to predict the cardiotoxic mode of action of DOX in patients. These results illustrate "antioxidant" properties of DOX, mediated by reduction of Mb(IV) to Mb(III), and cast doubts on lipid peroxidation as a causative mechanism of anthracycline-induced cardiotoxicity.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Doxorubicin/pharmacology , Heart/drug effects , Metmyoglobin/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Arachidonic Acid/metabolism , Doxorubicin/analogs & derivatives , Doxorubicin/toxicity , Free Radicals , Horses , Humans , Hydrogen Peroxide/metabolism , Inhibitory Concentration 50 , Iron/metabolism , Kinetics , Lipid Peroxidation/drug effects , Metmyoglobin/chemistry , Myocardium/metabolism , Oxidation-Reduction/drug effects , Statistics as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...