Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Pharmacol ; 155(1): 138-48, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18536744

ABSTRACT

BACKGROUND AND PURPOSE: The clinical utility of anthracycline antineoplastic drugs is limited by the risk of cardiotoxicity, which has been traditionally attributed to iron-mediated production of reactive oxygen species (ROS). EXPERIMENTAL APPROACH: The aims of this study were to examine the strongly lipophilic iron chelator, salicylaldehyde isonicotinoyl hydrazone (SIH), for its ability to protect rat isolated cardiomyocytes against the toxicity of daunorubicin (DAU) and to investigate the effects of SIH on DAU-induced inhibition of proliferation in a leukaemic cell line. Cell toxicity was measured by release of lactate dehydrogenase and staining with Hoechst 33342 or propidium iodide and lipid peroxidation by malonaldehyde formation. KEY RESULTS: SIH fully protected cardiomyocytes against model oxidative injury induced by hydrogen peroxide exposure. SIH also significantly but only partially and with no apparent dose-dependency, reduced DAU-induced cardiomyocyte death. However, the observed protection was not accompanied by decreased lipid peroxidation. In the HL-60 acute promyelocytic leukaemia cell line, SIH did not blunt the antiproliferative efficacy of DAU. Instead, at concentrations that reduced DAU toxicity to cardiomyocytes, SIH enhanced the tumoricidal action of DAU. CONCLUSIONS AND IMPLICATIONS: This study demonstrates that iron is most likely involved in anthracycline cardiotoxicity and that iron chelation has protective potential, but apparently through mechanism(s) other than by inhibition of ROS-induced injury. In addition to cardioprotection, iron chelation may have considerable potential to improve the therapeutic action of anthracyclines by enhancing their anticancer efficiency and this potential warrants further investigation.


Subject(s)
Aldehydes/pharmacology , Antibiotics, Antineoplastic/toxicity , Daunorubicin/toxicity , Hydrazones/pharmacology , Iron Chelating Agents/pharmacology , Leukemia, Promyelocytic, Acute/pathology , Myocytes, Cardiac/drug effects , Animals , Animals, Newborn , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytoprotection , Dose-Response Relationship, Drug , HL-60 Cells , Humans , Leukemia, Promyelocytic, Acute/metabolism , Lipid Peroxidation/drug effects , Malondialdehyde/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Rats , Rats, Wistar , Time Factors
2.
Cell Biol Toxicol ; 23(1): 39-47, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17063376

ABSTRACT

Doxorubicin is a widely used anthracycline anticancer agent. Its use may cause cardiomyopathy: in fact, the development of cumulative dose-related cardiotoxicity forms the major limitation of clinical doxorubicin use. We therefore searched for protective agents that combine iron-chelating and oxygen radical-scavenging properties. Moreover, any novel protector should not interfere with the cytostatic activity of doxorubicin. After extensive in vitro screening we found that flavonoids could serve this purpose. In particular 7-monohydroxyethylrutoside almost completely protected against the negative inotropic action of doxorubicin in the electrically paced mouse left atrium model. In vivo it gave full protection at 500 mg/kg intraperitoneally against the doxorubicin-induced ST-interval lengthening in the ECG. Moreover, this protector did not influence the antitumor effect of doxorubicin either in vitro using the human ovarian cell lines A2780 and OVCAR-3 and the human breast cancer cell line MCF-7 or in vivo in A2780 and OVCAR-3 subcutaneous xenografts in nude mice. Comparison of various iron chelators suggest that iron, in contrast to the general assumption, might not play a crucial role in the oxidative stress-induced toxicity of doxorubicin. Moreover, incubation of vascular endothelial cells with doxorubicin produced overexpression of adhesion molecules, which could be inhibited by 7-monohydroxyethylrutoside. From a study in human volunteers, we conclude that an intravenous dose of 1500 mg/m(2) of 7-monohydroxyethylrutoside is feasible and is safe to be investigated as protection against doxorubicin-induced cardiotoxicity.


Subject(s)
Cardiotonic Agents/pharmacology , Doxorubicin/toxicity , Flavonoids/pharmacology , Heart/drug effects , Animals , Antibiotics, Antineoplastic/toxicity , Cell Line, Tumor , Drug Evaluation, Preclinical/methods , Electrocardiography , Female , Heart/physiopathology , Humans , Hydroxyethylrutoside/analogs & derivatives , Hydroxyethylrutoside/pharmacology , In Vitro Techniques , Inflammation Mediators/metabolism , Iron/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Myocardial Contraction/drug effects , Superoxides/metabolism
3.
Br J Pharmacol ; 149(7): 920-30, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17031387

ABSTRACT

BACKGROUND AND PURPOSE: The anticancer drugs doxorubicin and bleomycin are well-known for their oxidative stress-mediated side effects in heart and lung, respectively. It is frequently suggested that iron is involved in doxorubicin and bleomycin toxicity. We set out to elucidate whether iron chelation prevents the oxidative stress-mediated toxicity of doxorubicin and bleomycin and whether it affects their antiproliferative/proapoptotic effects. EXPERIMENTAL APPROACH: Cell culture experiments were performed in A549 cells. Formation of hydroxyl radicals was measured in vitro by electron paramagnetic resonance (EPR). We investigated interactions between five iron chelators and the oxidative stress-inducing agents (doxorubicin, bleomycin and H(2)O(2)) by quantifying oxidative stress and cellular damage as TBARS formation, glutathione (GSH) consumption and lactic dehydrogenase (LDH) leakage. The antitumour/proapoptotic effects of doxorubicin and bleomycin were assessed by cell proliferation and caspase-3 activity assay. KEY RESULTS: All the tested chelators, except for monohydroxyethylrutoside (monoHER), prevented hydroxyl radical formation induced by H(2)O(2)/Fe(2+) in EPR studies. However, only salicylaldehyde isonicotinoyl hydrazone and deferoxamine protected intact A549 cells against H(2)O(2)/Fe(2+). Conversely, the chelators that decreased doxorubicin and bleomycin-induced oxidative stress and cellular damage (dexrazoxane, monoHER) were not able to protect against H(2)O(2)/Fe(2+). CONCLUSIONS AND IMPLICATIONS: We have shown that the ability to chelate iron as such is not the sole determinant of a compound protecting against doxorubicin or bleomycin-induced cytotoxicity. Our data challenge the putative role of iron and hydroxyl radicals in the oxidative stress-mediated cytotoxicity of doxorubicin and bleomycin and have implications for the development of new compounds to protects against this toxicity.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Doxorubicin/toxicity , Iron Chelating Agents/pharmacology , Iron Compounds/metabolism , Lung Neoplasms/metabolism , Oxidative Stress/drug effects , Aldehydes/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Deferoxamine/pharmacology , Electron Spin Resonance Spectroscopy , Free Radicals/chemistry , Humans , Hydrazones/pharmacology , Hydrogen Peroxide/chemistry , Iron/chemistry , Iron Chelating Agents/chemistry , Iron Compounds/chemistry , Isoniazid/analogs & derivatives , Isoniazid/pharmacology , Lipid Peroxidation/drug effects , Lung Neoplasms/pathology , Pyridoxal/analogs & derivatives , Pyridoxal/pharmacology , Razoxane/pharmacology , Time Factors
4.
Physiol Res ; 53(6): 683-91, 2004.
Article in English | MEDLINE | ID: mdl-15588138

ABSTRACT

A major obstacle to the therapeutic use of anthracyclines, highly effective anticancer agents, is the fact that their administration results in dose-dependent cardiomyopathy. According to the currently accepted hypothesis, anthracyclines injure the heart by generating oxygen free radicals. The ability of pyridoxal isonicotinoyl hydrazone (PIH) and salicylaldehyde isonicotinoyl hydrazone (SIH) -- new iron chelators -- to protect against peroxidation as well as their suitable biological, physical and chemical properties make the compounds promising candidates for pre-clinical and clinical studies. Activities of carbonyl reductase CR (1.1.1.184), dihydrodiol dehydrogenase DD2 (1.3.1.20), aldehyde reductase ALR1 (1.1.1.2) and P450 isoenzymes (CYP1A1, CYP1A2, CYP2B, CYP3A) involved in the metabolism of daunorubicin, doxorubicin and other drugs or xenobiotics were studied. Various concentrations of the chelators were used either alone or together with daunorubicin or doxorubicin for in vitro studies in isolated hepatocytes. A significant decrease of activity was observed for all enzymes only at PIH and SIH concentrations higher than those presumed to be used for therapy. The results show that PIH and SIH have no effect on the activities of the enzymes studied in vitro and allow us to believe that they will not interfere with the metabolism of co-administered drugs and other xenobiotics. Daunorubicin (Da) and doxorubicin (Dx) significantly reduce cytochrome P450 activity, but the addition of SIH and PIH chelators (50 microM) reverses the reduction and restores the activity to 70-90 % of the activity of relevant controls.


Subject(s)
Aldehydes/pharmacology , Anthracyclines/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Hepatocytes/metabolism , Hydrazones/pharmacology , Isoniazid/analogs & derivatives , Isoniazid/pharmacology , Oxidoreductases/metabolism , Pyridoxal/analogs & derivatives , Pyridoxal/pharmacology , Animals , Anthracyclines/metabolism , Antibiotics, Antineoplastic/metabolism , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cells, Cultured , Chelating Agents/pharmacology , Cytochrome P-450 Enzyme System/drug effects , Cytosol/drug effects , Cytosol/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Hepatocytes/drug effects , Male , Oxidoreductases/drug effects , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...